201
|
Kolkowski EC, Fernández MA, Pujol-Borrell R, Jaraquemada D. Human intestinal alphabeta IEL clones in celiac disease show reduced IL-10 synthesis and enhanced IL-2 production. Cell Immunol 2007; 244:1-9. [PMID: 17368439 DOI: 10.1016/j.cellimm.2007.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 01/13/2007] [Accepted: 01/18/2007] [Indexed: 01/14/2023]
Abstract
Celiac disease is a gluten-induced T-cell mediated autoimmune process that results in the destruction of the intestinal mucosa and is associated with an expansion of CD8(+) CD103(+) TCRalphabeta intraepithelial lymphocytes (IELs) in the damaged epithelium. The role of this IEL population in the pathology is unknown. The aim of this work was to compare the cytokine profile and the cytotoxicity pattern from CD8(+) IEL clones isolated from celiac (CD) and non-celiac (NCD) biopsies. We report that the number of IL-10 producing CD clones was significantly lower (26%) than that obtained from the NCD sample (62%). Instead, IL-2 was produced by more CD (44%) than NCD clones (26%). Cytotoxicity patterns against intestinal epithelial cell lines suggest different functional subsets of CD8(+) IELs. CD clones capable of high cytotoxicity produced IL-2 whereas most cytotoxic NCD IELs produced IL-10. This clonal analysis indicates that an impaired immune regulation in celiac mucosa may be partially attributed to the low generation of regulatory CD8(+) IELs that produce IL-10.
Collapse
Affiliation(s)
- Edgardo C Kolkowski
- Unitat d'Immunologia, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | | | |
Collapse
|
202
|
Abstract
Celiac disease is characterized by small-intestinal mucosal injury and nutrient malabsorption in genetically susceptible individuals in response to the dietary ingestion of wheat gluten and similar proteins in barley and rye. Disease pathogenesis involves interactions among environmental, genetic, and immunological factors. Although celiac disease is predicted by screening studies to affect approximately 1% of the population of the United States and is seen both in children and in adults, 10%-15% or fewer of these individuals have been diagnosed and treated. This article focuses on the role of adaptive and innate immune mechanisms in the pathogenesis of celiac disease and how current concepts of immunopathogenesis might provide alternative approaches for treating celiac disease.
Collapse
Affiliation(s)
- Martin F Kagnoff
- Department of Medicine, Laboratory of Mucosal Immunology, and Wm. K. Warren Medical Research Center for Celiac Disease, UCSD, La Jolla, California 92093-0623, USA.
| |
Collapse
|
203
|
Cinova J, Palová-Jelínková L, Smythies LE, Cerná M, Pecharová B, Dvorák M, Fruhauf P, Tlaskalová-Hogenová H, Smith PD, Tucková L. Gliadin peptides activate blood monocytes from patients with celiac disease. J Clin Immunol 2007; 27:201-9. [PMID: 17260166 DOI: 10.1007/s10875-006-9061-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 12/01/2006] [Indexed: 02/06/2023]
Abstract
To elucidate the role of innate immune responses in celiac disease, we investigated the effect of gliadin on blood monocytes from patients with celiac disease. Gliadin induced substantial TNF-alpha and IL-8 production by monocytes from patients with active celiac disease, lower levels by monocytes from patients with inactive celiac disease, and even lower levels by monocytes from healthy donors. In healthy donor monocytes gliadin induced IL-8 from monocytes expressing HLA-DQ2 and increased monocyte expression of the costimulatory molecules CD80 and CD86, the dendritic cell marker CD83, and the activation marker CD40. Gliadin also increased DNA binding activity of NF-kappaB p50 and p65 subunits in monocytes from celiac patients, and NF-kappaB inhibitors reduced both DNA binding activity and cytokine production. Thus, gliadin activation of HLA-DQ2(+) monocytes leading to chemokine and proinflammatory cytokine production may contribute to the host innate immune response in celiac disease.
Collapse
Affiliation(s)
- Jana Cinova
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Nilsson L, Kivling A, Jalmelid M, Fälth Magnusson K, Faresjö M. Combinations of common chronic paediatric diseases deviate the immune response in diverging directions. Clin Exp Immunol 2007; 146:433-42. [PMID: 17100762 PMCID: PMC1810401 DOI: 10.1111/j.1365-2249.2006.03228.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The cytokine pattern of T lymphocytes has not been characterized in children with combinations of paediatric immunological disorders. We describe cytokine secretion in children with type 1 diabetes, coeliac disease and allergy and combinations of two of these diseases after stimulation with 'disease-specific' antigens. Peripheral blood mononuclear cells (PBMC) were collected from 68 children with type 1 diabetes, allergy or coeliac disease, two of these diseases in combination or none of these diseases. Using the enzyme-linked immunospot (ELISPOT) technique, interferon (IFN)-gamma and interleukin (IL)-4 were analysed from fresh PBMC spontaneously and after in vitro stimulation with antigens associated with one or more of these diseases (insulin, gluten, birch and cat extract, beta-lactoglobulin, ovalbumin and phytohaemagglutinin) in order to divide T helper (Th)1- from Th2-like lymphocytes. Stimulation with birch and cat extract caused increased IL-4 secretion in allergic children. A low IFN-gamma response to insulin was found in type 1 diabetic children, whereas allergic children responded to insulin by increased IL-4 secretion. Children suffering from both type 1 diabetes (Th1-prone) and allergy (Th2-prone) reacted distinctly to general mitogen stimulation. Children suffering from two Th1-dominated diseases (type 1 diabetes and coeliac disease) showed hardly any response to either food or inhalation allergens. Our results indicate an important interplay between common immunological diseases in children. The combination of two Th1-deviated diseases is associated with a suppressed immune response, whereas a combination of Th1- and Th2-dominated diseases appears to increase the general immune response.
Collapse
Affiliation(s)
- L Nilsson
- Division of Paediatrics, Department of Molecular and Clinical Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | |
Collapse
|
205
|
León AJ, Garrote JA, Blanco-Quirós A, Calvo C, Fernández-Salazar L, Del Villar A, Barrera A, Arranz E. Interleukin 18 maintains a long-standing inflammation in coeliac disease patients. Clin Exp Immunol 2007; 146:479-85. [PMID: 17100768 PMCID: PMC1810422 DOI: 10.1111/j.1365-2249.2006.03239.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Dietary gluten induces an early response in the intestine of coeliac disease patients (CD), within a few hours, and this is driven by high levels of proinflammatory cytokines, including IFNgamma and IL-15, as has been thoroughly shown by gluten stimulation of biopsy explants. Our aim was to identify the immune mediators involved in the long-standing inflammation in untreated CD patients at diagnosis. mRNA and protein levels of TNFalpha, IL-12(p35), IL-12(p40), IL-15, IL-18 and IL-23(p19) were quantified in biopsies from active CD patients, CD patients on a gluten-free diet (GFD), healthy controls, and patients with non-CD inflammation and mild histological changes in the intestine. Biopsies from CD patients on a GFD were also stimulated in vitro with gliadin, and protein expression of IL-15 and IL-18 was analysed. Levels of IL-12 and IL-23 mRNA are nearly absent, and TNFalpha levels remain unchanged among different groups. Both the active and inactive forms of IL-18 protein have been found in all samples from active CD, and protein expression was only localized within the crypts. Levels of IL-15 mRNA remain unchanged, and protein expression, localized within the lamina propria, is found in a small number of samples. In vitro stimulation with gluten induces the expression of IL-15 and IL-18. In active CD, the early response following gluten intake characterized by high IFNgamma levels is driven by IL-18, and probably IL-15, and this alternates with periods of long-standing inflammation with moderate IFNgamma levels, maintained by IL-18 alone.
Collapse
Affiliation(s)
- A J León
- Department of Paediatrics and Immunology, and Institute of Biology and Molecular Genetics (IBGM), Universidad de ValladolidValladolid, Spain
| | - J A Garrote
- Department of Paediatrics and Immunology, and Institute of Biology and Molecular Genetics (IBGM), Universidad de ValladolidValladolid, Spain
- Research Unit, Hospital Clínico UniversitarioValladolid, Spain
| | - A Blanco-Quirós
- Department of Paediatrics and Immunology, and Institute of Biology and Molecular Genetics (IBGM), Universidad de ValladolidValladolid, Spain
| | - C Calvo
- Department of Paediatrics and Immunology, and Institute of Biology and Molecular Genetics (IBGM), Universidad de ValladolidValladolid, Spain
- Paediatric Gastroenterology Clinics, Hospital Clínico UniversitarioValladolid, Spain
| | | | - A Del Villar
- Pathology, Hospital Universitario Rio-HortegaValladolid, Spain
| | - A Barrera
- General and Digestive Surgery Units, Hospital Universitario Rio-HortegaValladolid, Spain
| | - E Arranz
- Department of Paediatrics and Immunology, and Institute of Biology and Molecular Genetics (IBGM), Universidad de ValladolidValladolid, Spain
| |
Collapse
|
206
|
Abstract
Celiac disease (CD) is an (auto)immunologically mediated intestinal intolerance against proteins from wheat (gluten) and related cereal proteins. Tissue transglutaminase (tTG) has been identified as the autoantigen in CD. Although ultimate diagnosis is based on histological analysis of small intestinal mucosa obtained via tissue biopsy, assessment of autoantibodies can provide substantial help in the evaluation of CD. Gliadin antibodies are directed against the native disease-provoking cereal proteins. Despite their initial usefulness, these antibodies have lost diagnostic importance due to their poor specificity and sensitivity as CD markers. Recently, it was found, however, that gliadin antibodies from sera of patients with active CD preferentially recognized deamidated gliadin peptides. The use of deamidated gliadin peptides in immunoassays has significantly improved the usefulness of gliadin antibodies in diagnosis of CD to that observed with autoantibody assay methods (endomysium antibodies, antibodies against tTG). The antibody epitopes (B-cell epitopes) reflect substrate specificity of tTG and resemble peptide sequences known to be strongly T-cell stimulatory (T-cell epitopes) in CD. The assay applying deamidated gliadin peptides measures a new species of antibodies, which is different from conventional gliadin antibodies as well as from autoantibodies and will likely provide new information on pathophysiological mechanisms of CD.
Collapse
Affiliation(s)
- Thomas Mothes
- Institute for Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital and Medical Faculty of the University, Leipzig, Germany
| |
Collapse
|
207
|
Pizzuti D, Buda A, D'Odorico A, D'Incà R, Chiarelli S, Curioni A, Martines D. Lack of intestinal mucosal toxicity of Triticum monococcum in celiac disease patients. Scand J Gastroenterol 2006; 41:1305-11. [PMID: 17060124 DOI: 10.1080/00365520600699983] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The treatment of celiac disease is based on lifelong withdrawal of foods containing gluten. Unfortunately, compliance with a gluten-free diet has proved poor in many patients (mainly due to its low palatability), emphasizing the need for cereal varieties that are not toxic for celiac patients. In evolutionary terms, Triticum monococcum is the oldest and most primitive cultivated wheat. The aim of this study was to evaluate the toxicity of T. monococcum on small intestinal mucosa, using an in vitro organ culture system. MATERIAL AND METHODS Distal duodenum biopsies of 12 treated celiac patients and 17 control subjects were cultured for 24 h with T. aestivum (bread) gliadin (1 mg/ml) or with T. monococcum gliadin (1 mg/ml). Biopsies cultured with medium alone served as controls. Each biopsy was used for conventional histological examination and for immunohistochemical detection of CD3 + intraepithelial lymphocytes (IELs) and HLA-DR. Secreted cytokine protein interferon-gamma (IFN-gamma) was measured in the culture supernatant using an enzyme-linked immunoadsorbent assay. RESULTS Significant morphological changes, HLA-DR overexpression in the crypt epithelium and an increased number of CD3 + IELs, found after bread gliadin exposure, were not observed in celiac biopsies cultured with T. monococcum gliadin. In contrast, with bread gliadin, there was no significant IFN-gamma response after culture with monococcum gliadin. Similarly, biopsies from normal controls did not respond to bread or monococcum gliadin stimulation. CONCLUSIONS These data show a lack of toxicity of T. monococcum gliadin in an in vitro organ culture system, suggesting new dietary opportunities for celiac patients.
Collapse
Affiliation(s)
- Daniela Pizzuti
- Department of Surgical and Gastroenterological Sciences, Padua University, Italy.
| | | | | | | | | | | | | |
Collapse
|
208
|
Ménard S, Candalh C, Ben Ahmed M, Rakotobe S, Gaboriau-Routhiau V, Cerf-Bensussan N, Heyman M. Stimulation of immunity without alteration of oral tolerance in mice fed with heat-treated fermented infant formula. J Pediatr Gastroenterol Nutr 2006; 43:451-8. [PMID: 17033519 DOI: 10.1097/01.mpg.0000239738.71864.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Little information is available on the properties of fermented milk formula intended to healthy infants. This study analyzes the effect of long-term ingestion of a heat-treated, fermented milk formula on the development of oral tolerance or systemic immune response to soluble antigens in mice. MATERIALS AND METHODS The C3H/HeN mice, fed with a heat-treated fermented (Bifidobacterium breve C50 and Streptococcus thermophilus 065) infant formula (htFF) or a matched control diet (control), were immunized with ovalbumin (OVA) with or without gavage of 20 mg OVA to induce tolerance or immunity, respectively. Systemic and local anti-OVA immune responses and intestinal barrier function were measured after 5 to 6 weeks. RESULTS Oral tolerance to OVA developed similarly in htFF- and control-fed mice, attested to by the downregulation of OVA-specific immunoglobulin (Ig) G and IgE after oral OVA administration. In contrast, immunization with OVA led to significantly higher titers in htFF-fed mice than in control-fed mice (log2 IgG titers, 16.45 +/- 1.24 and 15.46 +/- 0.79, respectively; P = 0.012). Jejunal interferon gamma, interleukin 12p40 and interleukin 10 expressions were significantly higher in tolerized mice fed with htFF compared with those fed with the control diet. Mucosal to serosal intact horseradish peroxidase fluxes were lower in htFF-fed mice than in control-fed mice (39 +/- 8 and 118 +/- 38 ng/h x cm2, respectively; P < 0.0001), indicating that the htFF diet reinforces intestinal barrier capacity to macromolecules. CONCLUSIONS In mice, htFF strengthens intestinal barrier and enhances systemic immune responses to antigens without interfering with the development of oral tolerance, suggesting a potential beneficial effect in host defence and vaccination.
Collapse
|
209
|
Daum S, Foss HD, Schuppan D, Riecken EO, Zeitz M, Ullrich R. Synthesis of collagen I in collagenous sprue. Clin Gastroenterol Hepatol 2006; 4:1232-6. [PMID: 16979955 DOI: 10.1016/j.cgh.2006.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Collagenous sprue (CS) is a rare severe malabsorption syndrome of unknown etiology, characterized by villus atrophy and a broad subepithelial collagen band. We studied the expression patterns of genes involved in fibrogenesis and fibrolysis and analyzed the composition of the subepithelial collagen band in CS compared with untreated celiac diseases (CDs). METHODS Duodenal biopsies from 2 patients with CS were hybridized with (35)S-labeled RNA probes for procollagen alpha1(I), matrix metalloproteinases (MMPs)-1 and -3, and tissue inhibitor of MMP-1 (TIMP-1). Numbers of positive subepithelial and lamina propria cells and grain density per cell were determined microscopically. Immunohistochemistry for collagens I, III, IV, XIV, and tenascin was performed by the immunoalkaline phosphatase method. Findings were compared with earlier data from patients with untreated CD. RESULTS Numbers of cells expressing procollagen I, MMP-1, and MMP-3 mRNA were similar, but subepithelial procollagen I transcripts per cell were significantly increased (42 and 38 vs 18 [13-23]; P < .05) in CS compared with CD, whereas cellular transcript densities for MMP-1 and MMP-3 mRNA were similar. In addition, the number of TIMP-1 mRNA-positive cells was higher in CS compared with CD (32 and 25 vs 16.5 [4-25]; P < .05). The subepithelial collagenous bands stained for collagen I, collagen III, and tenascin. CONCLUSIONS The prominent subepithelial matrix deposition in CS is due to increased expression of the main fibrogenic genes (procollagen I and TIMP-1) by myofibroblastic cells, whereas expression of fibrolytic MMPs remains unaltered.
Collapse
Affiliation(s)
- Severin Daum
- Department of Medicine I, Gastroenterology, Infectious Diseases and Rheumatology, Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
210
|
Biagi F, Campanella J, Laforenza U, Gastaldi G, Tritto S, Grazioli M, Villanacci V, Corazza GR. Transglutaminase 2 in the enterocytes is coeliac specific and gluten dependent. Dig Liver Dis 2006; 38:652-8. [PMID: 16916632 DOI: 10.1016/j.dld.2006.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 04/24/2006] [Accepted: 05/22/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tissue transglutaminase, the coeliac autoantigen, was shown to localise in the enterocytes of coeliac patients and controls. It was speculated that surface tissue transglutaminase has a role in the pathogenesis of coeliac disease. AIMS To study localisation of tissue transglutaminase in different stages of coeliac disease and other enteropathies with and without villous flattening. METHODS Immunofluorescent and immunoblotting assays were used. Duodenal cryostat sections from 23 coeliac patients (10 untreated, 8 treated, 5 potential) and 18 controls (2 autoimmune enteropathy and 16 normal duodenal mucosa) were incubated with an anti-tissue transglutaminase monoclonal antibody. Slides were blindly examined. RESULTS The immunofluorescent assay showed that monoclonal antibody localised in the subepithelial layer, in the lamina propria, and in the pericryptal connective tissue of all samples. It also bound to surface enterocytes in 8/10 untreated, 1/8 treated, and 3/5 potential coeliac patients. None of the controls showed an epithelial distribution of tissue transglutaminase. Immunoblotting experiments performed in enterocytes freshly isolated from duodenal biopsy confirmed these findings. CONCLUSION Epithelial distribution of tissue transglutaminase is specific for coeliac disease rather than due to a non-specific mucosal inflammation. Analysis of different stages of coeliac disease suggests that the epithelial distribution of tissue transglutaminase is gluten dependent.
Collapse
Affiliation(s)
- F Biagi
- 1st Department of Internal Medicine, IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi, 19, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Branski D, Fasano A, Troncone R. Latest developments in the pathogenesis and treatment of celiac disease. J Pediatr 2006; 149:295-300. [PMID: 16939736 DOI: 10.1016/j.jpeds.2006.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 05/05/2006] [Accepted: 06/05/2006] [Indexed: 01/01/2023]
Affiliation(s)
- David Branski
- Department of Pediatrics, Hadassah University Hospitals, Hadassah Medical Organization, 91120 Jerusalem, Israel.
| | | | | |
Collapse
|
212
|
Rakoff-Nahoum S, Hao L, Medzhitov R. Role of toll-like receptors in spontaneous commensal-dependent colitis. Immunity 2006; 25:319-29. [PMID: 16879997 DOI: 10.1016/j.immuni.2006.06.010] [Citation(s) in RCA: 299] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Revised: 04/26/2006] [Accepted: 06/12/2006] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is thought to result from a dysregulated interaction between the host immune system and its commensal microflora. Heterogeneity of disease susceptibility in humans and rodents suggest that multiple mechanisms are responsible for the etiology of IBD. In particular, deficiencies in anti-inflammatory and immune-suppressive mechanisms play an important role in the development of IBD. However, it is unknown how the indigenous microflora stimulates the immune system and how this response is regulated. To address these questions, we investigated the role of Toll-like receptor (TLR) signaling in the development of spontaneous, commensal-dependent colitis in interleukin (IL)-2- and IL-10-deficient mice. We report that colitis was dependent on TLR signaling in Il10(-/-) mice. In contrast, Il2(-/-) mice developed intestinal inflammation in the absence of TLR signaling pathways. These results demonstrate a differential role of innate immune recognition by TLRs in the development of commensal-dependent colitis.
Collapse
Affiliation(s)
- Seth Rakoff-Nahoum
- Howard Hughes Medical Institute and Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | |
Collapse
|
213
|
Agardh D, Lynch K, Brundin C, Ivarsson SA, Lernmark A, Cilio CM. Reduction of tissue transglutaminase autoantibody levels by gluten-free diet is associated with changes in subsets of peripheral blood lymphocytes in children with newly diagnosed coeliac disease. Clin Exp Immunol 2006; 144:67-75. [PMID: 16542367 PMCID: PMC1809644 DOI: 10.1111/j.1365-2249.2006.03036.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tissue transglutaminase (tTG) autoantibodies decline after gluten-free diet in patients with coeliac disease. We tested the hypothesis that gluten-free diet-induced change in tTG autoantibody levels affects subsets of peripheral blood lymphocytes. Peripheral blood was obtained from 20 children with biopsy-proven active coeliac disease. Gluten-free diet was initiated and the children examined again after three and six months. tTG autoantibodies were measured in radioligand binding assays and lymphocyte subsets by flow cytometry. IgA-tTG levels at diagnosis, 2204 U/ml (median, range 113-24990), were reduced over six months to 76 U/ml (median, range 1-1261) (P < 0.001). At six months, 12/20 (60%) children had reduced their IgA-tTG levels to < 100 U/ml and these children showed a decrease in B cells (mean change -3.8%, P = 0.014), CD4+ T cells (mean -4.32%, P = 0.011) and CD4+ T cells expressing CD25high (mean change -0.62%, P = 0.036). In contrast, the CD4+CD25(high)CCR4+ T cell population increased during the same period (mean change 11.5%, P = 0.0036). The decline in IgA-tTG levels correlated to the decrease in B cells (r = 0.56, P = 0.01), CD4+ T cells (r = 0.66, P = 0.004) as well as CD4+CD25high T cells (r = 0.59, P = 0.01). A negative correlation was found between the decline in IgA-tTG and CD4+CD25high T cells expressing CD45RO (r = -0.49, P = 0.03) and CCR4 (r = -0.54, P = 0.01). This is the first observational study on the effect of gluten-free diet on concurrent changes of tTG autoantibodies and specific peripheral blood lymphocyte subsets. Our data suggest that flow cytometry may be a useful complement to tTG autoantibodies when studying the effects of gluten-free diet in children with coeliac disease.
Collapse
Affiliation(s)
- D Agardh
- Unit of Diabetes and Coeliac Disease, Department of Clinical Sciences/Paediatrics, Lund University, Malmö, Sweden
| | | | | | | | | | | |
Collapse
|
214
|
Tagkalidis PP, Gibson PR, Bhathal PS. Microscopic colitis demonstrates a T helper cell type 1 mucosal cytokine profile. J Clin Pathol 2006; 60:382-7. [PMID: 16775121 PMCID: PMC2001106 DOI: 10.1136/jcp.2005.036376] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Microscopic colitis (MC) is an inflammatory disorder of unknown aetiology. AIM To characterise the mucosal cytokine profile of MC, with a view to understanding its potential pathogenic mechanisms. METHODS Cytokine profiles of mucosal biopse specimens taken at flexible sigmoidoscopy from 18 patients (8 with lymphocytic colitis and 10 with collagenous colitis) were analysed using real-time reverse transcriptase-PCR, in comparison with those from 13 aged-matched controls with diarrhoea-predominant irritable bowel syndrome. Biopsy specimens from six patients with histologically documented remission were available for comparative analysis. Biopsy specimens were also taken to determine the cellular expression of cytokine and cytokine-related proteins using immunohistochemistry. RESULTS Mucosal mRNA levels were 100 times greater for interferon (IFN)gamma and interleukin (IL) 15, 60 times greater for tumour necrosis factor alpha, and 35 times greater for inducible nitric oxide synthase in MC compared with controls. Apart from a trend for increased levels of IL10, levels of other T helper cell type 2 (T(H)2) cytokines including IL2 and IL4 were too low to be accurately quantified. Mucosal IFNgamma mRNA levels correlated with the degree of diarrhoea, and returned to normal in remission. The immunohistochemical expression of cell junction proteins E-cadherin and ZO-1 was reduced in active disease. No differences were noted between lymphocytic and collagenous colitis for any of the above parameters. CONCLUSIONS MC demonstrates a T(H)1 mucosal cytokine profile with IFNgamma as the predominantly upregulated cytokine, with concurrent induction of nitric oxide synthase and down regulation of IFNgamma-related cell junction proteins. This pattern is similar to that in coeliac disease and suggests that it might represent a response to a luminal antigen.
Collapse
Affiliation(s)
- Peter P Tagkalidis
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | |
Collapse
|
215
|
Srinivasan U, Jones E, Carolan J, Feighery C. Immunohistochemical analysis of coeliac mucosa following ingestion of oats. Clin Exp Immunol 2006; 144:197-203. [PMID: 16634791 PMCID: PMC1809658 DOI: 10.1111/j.1365-2249.2006.03052.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2006] [Indexed: 01/25/2023] Open
Abstract
There is now considerable clinical evidence that oats do not activate coeliac disease. Nonetheless, a reluctance to include oats in the gluten-free diet remains. Because gluten-induced damage is accompanied by activation of the gastrointestinal immune system, the purpose of this study was to investigate if similar changes were induced by oats ingestion. Small intestinal histological sections from 10 patients who ingested 50 g of oats daily for 3 months were investigated for possible evidence of immune activation. Tissue obtained before and after oats challenge was stained with a series of antibodies directed against the following molecules: human leucocyte antigen D-related (HLA-DR), Ki-67, CD25, CD54 [intercellular adhesion molecule 1 (ICAM-1)] and mast cell tryptase. None of the patients developed clinical or laboratory evidence of adverse effects. The distribution of intestinal HLA-DR expression was not affected by oats ingestion and the crypt epithelium remained unstained. In the pre-oats biopsies, the percentage of Ki-67 positive enterocytes, 29.5 +/- 6.9 [95% confidence interval (CI) 13.9-45.0] did not differ significantly from that found in post-oats biopsies, 41.2 +/- 3.7 (95% CI, 32.8-49.6), P = 0.19, not significant. Furthermore, oats ingestion did not alter the number of CD25 positive and tryptase positive cells. Finally, the distribution and intensity of ICAM-1 staining was unchanged by dietary oats. In summary, detailed immunohistological studies of biopsies from patients ingesting oats for 3 months did not reveal evidence of immune activation. Together with other reported findings, this study strengthens the view that oats can be included safely in the diet of gluten sensitive patients.
Collapse
Affiliation(s)
- U Srinivasan
- Department of Immunology, Trinity College Dublin and St James's Hospital, Dublin 8, Ireland
| | | | | | | |
Collapse
|
216
|
Bracken SC, Kilmartin C, Wieser H, Jackson J, Feighery C. Barley and rye prolamins induce an mRNA interferon-gamma response in coeliac mucosa. Aliment Pharmacol Ther 2006; 23:1307-14. [PMID: 16629935 DOI: 10.1111/j.1365-2036.2006.02876.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND In coeliac disease, wheat, barley and rye are traditionally excluded in the gluten-free diet. However, few studies have examined the small intestinal immune response to barley and rye. AIM To investigate the immunogenicity of barley and rye prolamins (hordein and secalin respectively) in comparison with wheat gliadin. METHODS Duodenal biopsies from 22 coeliac patients and 23 disease controls were cultured for 4 h with gliadin, hordein or secalin and compared with culture medium alone. Proinflammatory cytokines, interferon-gamma and interleukin-2, were quantified by TaqMan polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS Hordein caused the greatest increase in interferon-gamma mRNA in coeliac patients (median: 3.3-fold) in comparison with control subjects (median: 0.28-fold, P < 0.085). Secalin and gliadin induced similar levels of interferon-gamma mRNA with median fold-changes of 3.4 and 2.8, respectively, in coeliac patients in comparison with 1.6- and 1.1-fold increases in control subjects (P < 0.294 and P < 0.105, respectively). The median fold-changes for interleukin-2 mRNA did not differ between coeliac patients and controls. Cytokine protein was not upregulated. CONCLUSION The findings of this study provide evidence that barley and rye cause immune activation in the mucosa of coeliac patients and support the practice that barley and rye should be excluded from the gluten-free diet.
Collapse
Affiliation(s)
- S C Bracken
- Department of Immunology, Institute of Molecular Medicine, Trinity College Centre for Health Sciences, St James's Hospital, Dublin, Ireland
| | | | | | | | | |
Collapse
|
217
|
Brandtzaeg P. The changing immunological paradigm in coeliac disease. Immunol Lett 2006; 105:127-39. [PMID: 16647763 DOI: 10.1016/j.imlet.2006.03.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Revised: 03/13/2006] [Accepted: 03/15/2006] [Indexed: 02/06/2023]
Abstract
When coeliac disease is referred to as an inflammatory disorder, this may detract from its true nature. Activation of innate and adaptive immunity takes place in the mucosal lesion, but the tissue reaction is not that of classical inflammation. In fact, coeliac disease contrasts strikingly with typical inflammatory bowel disorders such as ulcerative colitis and Crohn's disease. The coeliac lesion apparently reflects, in the main, immune-driven remodelling of mucosal architecture with only a minor inflammatory component - initially most likely resulting from innate signals. Complement split products might be one of several potential initial hits that lead to activation of lamina propria and epithelial cells with release of mediators such as interleukin-15. This cytokine appears to stimulate potentially pathogenic intraepithelial lymphocytes. In genetically susceptible individuals, such early innate events could turn into persistent pathogenic signalling with subsequent adaptive cellular and humoral immunopathology resulting in a chronic lesion. Nevertheless, mucosal homeostasis is surprisingly well preserved as signified by the remarkable dominance of plasma cells that produce dimeric immunoglobulin A as a basis for enhanced secretory immunity. This shows that the microvascular endothelium in the lesion largely maintains its 'gatekeeper' function for mucosal immune cells - in striking contrast to the 'promiscuous' situation in inflammatory bowel disease. Altogether, a two-signal model is emerging for the pathogenesis of coeliac disease - signal 1 generated by innate immunity and signal 2 by adaptive immunity. Hence, there is currently an increased focus on immune activation in the epithelial compartment rather than on changes in the microvasculature as a basis for classical inflammation.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Institute and Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Centre, N-0027 Oslo, Norway.
| |
Collapse
|
218
|
Núñez C, Alecsandru D, Varadé J, Polanco I, Maluenda C, Fernández-Arquero M, de la Concha EG, Urcelay E, Martínez A. Interleukin-10 haplotypes in Celiac Disease in the Spanish population. BMC MEDICAL GENETICS 2006; 7:32. [PMID: 16579847 PMCID: PMC1481547 DOI: 10.1186/1471-2350-7-32] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Accepted: 03/31/2006] [Indexed: 11/10/2022]
Abstract
Background Celiac disease (CD) is a chronic disorder characterized by a pathological inflammatory response after exposure to gluten in genetically susceptible individuals. The HLA complex accounts for less than half of the genetic component of the disease, and additional genes must be implicated. Interleukin-10 (IL-10) is an important regulator of mucosal immunity, and several reports have described alterations of IL-10 levels in celiac patients. The IL-10 gene is located on chromosome 1, and its promoter carries several single nucleotide polymorphisms (SNPs) and microsatellites which have been associated to production levels. Our aim was to study the role of those polymorphisms in susceptibility to CD in our population. Methods A case-control and a familial study were performed. Positions -1082, -819 and -592 of the IL-10 promoter were typed by TaqMan and allele specific PCR. IL10R and IL10G microsatellites were amplified with labelled primers, and they were subsequently run on an automatic sequencer. In this study 446 patients and 573 controls were included, all of them white Spaniards. Extended haplotypes encompassing microsatellites and SNPs were obtained in families and estimated in controls by the Expectation-Maximization algorithm. Results No significant associations after Bonferroni correction were observed in the SNPs or any of the microsatellites. Stratification by HLA-DQ2 (DQA1*0501-DQB1*02) status did not alter the results. When extended haplotypes were analyzed, no differences were apparent either. Conclusion The IL-10 polymorphisms studied are not associated with celiac disease. Our data suggest that the IL-10 alteration seen in patients may be more consequence than cause of the disease.
Collapse
Affiliation(s)
- Concepción Núñez
- Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| | - Diana Alecsandru
- Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| | - Jezabel Varadé
- Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| | - Isabel Polanco
- Department of Paediatric Gastroenterology, Hospital La Paz, Madrid, Spain
| | - Carlos Maluenda
- Department of Paediatrics, Hospital Clínico San Carlos, Madrid, Spain
| | | | | | - Elena Urcelay
- Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| | - Alfonso Martínez
- Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
219
|
Hischenhuber C, Crevel R, Jarry B, Mäki M, Moneret-Vautrin DA, Romano A, Troncone R, Ward R. Review article: safe amounts of gluten for patients with wheat allergy or coeliac disease. Aliment Pharmacol Ther 2006; 23:559-75. [PMID: 16480395 DOI: 10.1111/j.1365-2036.2006.02768.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
For both wheat allergy and coeliac disease the dietary avoidance of wheat and other gluten-containing cereals is the only effective treatment. Estimation of the maximum tolerated amount of gluten for susceptible individuals would support effective management of their disease. Literature was reviewed to evaluate whether an upper limit for gluten content in food, which would be safe for sufferers from both diseases, could be identified. When setting gluten limits for coeliac disease sufferers, the overall potential daily intake should be considered, while for wheat allergy limits should be based on single servings. For coeliac disease sufferers this limit should lie between 10 and 100 mg daily intake. For wheat allergy, lowest eliciting doses for children lie in the lower milligram range, while for adults they are most significantly higher. Gliadins (part of the gluten proteins) not only trigger coeliac disease, but are also major allergens in wheat allergy. Therefore, measurement of gliadins with validated enzyme-linked immunosorbent assay methods provides an appropriate marker for assessing gluten and/or wheat protein contents in food. Available data suggest that a maximum gluten content for 'gluten-free' foods could be set, which protects both wheat allergy sufferers and coeliac patients.
Collapse
|
220
|
Barisani D, Ceroni S, Meneveri R, Cesana BM, Bardella MT. IL-10 polymorphisms are associated with early-onset celiac disease and severe mucosal damage in patients of Caucasian origin. Genet Med 2006; 8:169-174. [PMID: 16540751 DOI: 10.1097/01.gim.0000204464.87540.39] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Polymorphisms in cytokine genes can determine their level of expression and affect the phenotypic expression of immunomediated diseases, such as celiac disease (CD). We thus evaluated cytokine polymorphism prevalence in CD patients and population controls, and assessed the correlation between specific polymorphisms and celiacs' clinical characteristics. METHODS 155 CD patients and 202 population controls were enrolled in the study. Cytokine polymorphisms (TNFalpha -308 and -238, IL-1beta -511 and +3954, IL-1b RN + 2018, IL-6 -174, IL-10 -1082, -819 and -592, TGFbeta1 + 29 and +74, IFN-gamma + 874) and HLA class II alleles were determined by sequence-specific primer polymerase chain reaction or restriction fragment length polymorphism analysis. Duodenal histology was classified using Marsh's criteria. Variables significantly associated with CD patients' characteristics were identified by multivariate logistic regression analysis. RESULTS A significantly higher frequency of -308 TNFalpha polymorphism was observed in CD patients compared to the entire population control group, although no difference was detected when population controls were stratified according to HLA class II. Among celiacs, early onset of the disease (< or =2 year old) and the presence of a severe intestinal lesion (Marsh 3C) were significantly associated with the -1082 A/A IL-10 genotype which corresponds to a low producer phenotype (OR 3.28, 95% CI 1.49-7.19 and OR 2.60, 95% CI 1.04-6.49, respectively). CONCLUSION The association between IL-10 genotypes and both histological severity at diagnosis and age of onset could be related to an alteration in cytokine balance, and supports the idea that the various clinical manifestations of the disease could be determined by a different genetic background.
Collapse
Affiliation(s)
- Donatella Barisani
- Department of Experimental, Environmental Medicine and Medical Biotechnology, University of Milano Bicocca, Monza, Milan
| | | | | | | | | |
Collapse
|
221
|
Abstract
Celiac disease (CD) is the most common food-sensitive enteropathy in humans and is caused by the lack of immune tolerance (oral tolerance) to gluten. The identification of gluten-specific T cells in the lamina propria of celiacs and the strong association with HLA-DQ2 and -DQ8 genes support a central role of CD4(+) T cells in CD pathogenesis. Studies focused on the modulation of autoimmunity in different experimental models highlighted possible immune therapeutic protocols useful also for the management of CD. On the basis of these observations, a series of strategies have been designed: some of them are based on the identification of immunogenic epitopes and their suppression via enzymatic treatment or by using peptide analogues; others rely on the delivery of unmodified antigen through the nasal route or coadministered with downregulatory cytokines. studies are generally early stage but encouraging in paving a way for an alternative treatment for celiac disease.
Collapse
Affiliation(s)
- Mauro Rossi
- Istituto di Scienze dell'Alimentazione, CNR, Avellino, Italy.
| | | | | |
Collapse
|
222
|
Monteleone I, Monteleone G, Fina D, Caruso R, Petruzziello C, Calabrese E, Biancone L, Pallone F. A functional role of flip in conferring resistance of Crohn's disease lamina propria lymphocytes to FAS-mediated apoptosis. Gastroenterology 2006; 130:389-97. [PMID: 16472594 DOI: 10.1053/j.gastro.2005.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2004] [Accepted: 10/19/2005] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS There is evidence that, in Crohn's disease (CD), lamina propria T lymphocytes (LPLs) are resistant to FAS-mediated apoptosis and that this defect contributes to the mucosal T-cell accumulation. In this study we examined the functional role of Flip, a Flice inhibitor protein, in the resistance of CD LPL to FAS-mediated apoptosis. METHODS Biopsy specimens and LPLs were taken from CD and ulcerative colitis (UC) patients and normal controls and analyzed for Flip by Western blotting. We also examined whether inhibition of Flip by antisense oligonucleotide restored the susceptibility of CD LPLs to FAS-induced apoptosis. LPL apoptosis was assessed by flow cytometry. RESULTS After FAS stimulation, the rate of apoptosis of CD3+ LPLs was higher in normal controls and patients with UC than in patients with CD. Enhanced expression of both long and short Flip isoforms was seen in biopsy specimens and purified CD3+ and CD45RO+ LPLs of CD patients in comparison with UC patients and normal controls. No increase in Flip was documented in untreated celiac disease mucosa, thus suggesting the possibility that induction of Flip in the gut does not simply rely on the ongoing inflammation. Finally, we showed that inhibition of Flip by antisense oligonucleotide reverted the resistance of CD LPLs to FAS-induced apoptosis. CONCLUSIONS Data suggest a role for Flip in the resistance of CD LPLs to FAS-mediated apoptosis.
Collapse
Affiliation(s)
- Ivan Monteleone
- Dipartimento di Medicina Interna e Centro di Eccellenza per lo Studio Delle Malattie Complesse e Multifattoriali, Università Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
223
|
Palová-Jelínková L, Rozková D, Pecharová B, Bártová J, Sedivá A, Tlaskalová-Hogenová H, Spísek R, Tucková L. Gliadin fragments induce phenotypic and functional maturation of human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2006; 175:7038-45. [PMID: 16272365 DOI: 10.4049/jimmunol.175.10.7038] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Celiac disease is a chronic inflammatory disease developing in genetically predisposed individuals. Ingested gliadin, the triggering agent of the disease, can cross the epithelial barrier and elicit a harmful T cell-mediated immune response. Dendritic cells (DC) are supposed to play a pivotal role in shaping the immune response. The direction of the immune response toward immunity or tolerance depends on the stage of maturation and the functional properties of the DC. DC become fully functional APC upon maturation by various stimuli. We investigated the effect of a peptic digest of gliadin on the maturation of human monocyte-derived DC. Stimulation of cells with gliadin, in contrast with other tested food proteins, led to enhanced expression of maturation markers (CD80, CD83, CD86, and HLA-DR molecules) and increased secretion of chemokines and cytokines (mainly of IL-6, IL-8, IL-10, TNF-alpha, growth-related oncogene, MCP-1, MCP-2, macrophage-derived chemokine, and RANTES). Maturation was accompanied by a greater capacity to stimulate proliferation of allogeneic T cells and significantly reduced endocytic activity. Furthermore, gliadin-induced phosphorylation of members of three MAPK families (ERK1/2, JNK, and p38 MAPK) was demonstrated. The largest contribution of p38 MAPK was confirmed using its inhibitor SB203580, which markedly down-regulated the gliadin-triggered up-regulation of maturation markers and cytokine production. Gliadin treatment also resulted in increased NF-kappaB/DNA binding activity of p50 and p65 subunits. Taken together, gliadin peptides can contribute to overcoming the stage of unresponsiveness of immature DC by inducing phenotypic and functional DC maturation, resulting in more efficient processing and presentation of gliadin peptides to specific T lymphocytes.
Collapse
|
224
|
Senger S, Maurano F, Mazzeo MF, Gaita M, Fierro O, David CS, Troncone R, Auricchio S, Siciliano RA, Rossi M. Identification of Immunodominant Epitopes of α-Gliadin in HLA-DQ8 Transgenic Mice following Oral Immunization. THE JOURNAL OF IMMUNOLOGY 2005; 175:8087-95. [PMID: 16339546 DOI: 10.4049/jimmunol.175.12.8087] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Celiac disease, triggered by wheat gliadin and related prolamins from barley and rye, is characterized by a strong association with HLA-DQ2 and HLA-DQ8 genes. Gliadin is a mixture of many proteins that makes difficult the identification of major immunodominant epitopes. To address this issue, we expressed in Escherichia coli a recombinant alpha-gliadin (r-alpha-gliadin) showing the most conserved sequence among the fraction of alpha-gliadins. HLA-DQ8 mice, on a gluten-free diet, were intragastrically immunized with a chymotryptic digest of r-alpha-gliadin along with cholera toxin as adjuvant. Spleen and mesenteric lymph node T cell responses were analyzed for in vitro proliferative assay using a panel of synthetic peptides encompassing the entire sequence of r-alpha-gliadin. Two immunodominant epitopes corresponding to peptide p13 (aa 120-139) and p23 (aa 220-239) were identified. The response was restricted to DQ and mediated by CD4+ T cells. In vitro tissue transglutaminase deamidation of both peptides did not increase the response; furthermore, tissue transglutaminase catalyzed extensive deamidation in vitro along the entire r-alpha-gliadin molecule, but failed to elicit new immunogenic determinants. Surprisingly, the analysis of the cytokine profile showed that both deamidated and native peptides induced preferentially IFN-gamma secretion, despite the use of cholera toxin, a mucosal adjuvant that normally induces a Th2 response to bystander Ags. Taken together, these data suggest that, in this model of gluten hypersensitivity, deamidation is not a prerequisite for the initiation of gluten responses.
Collapse
Affiliation(s)
- Stefania Senger
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche, Avellino, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Ráki M, Molberg O, Tollefsen S, Lundin KEA, Sollid LM. The effects of atorvastatin on gluten-induced intestinal T cell responses in coeliac disease. Clin Exp Immunol 2005; 142:333-40. [PMID: 16232221 PMCID: PMC1809523 DOI: 10.1111/j.1365-2249.2005.02915.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2005] [Indexed: 01/17/2023] Open
Abstract
Various experimental models suggest that the cholesterol-lowering drugs statins may also modulate immune responses. Cellular level studies on human disorders are needed, however, to provide a rational basis for clinical testing of statins as immune therapy. Coeliac disease, a chronic small intestinal inflammation driven by HLA-DQ2 restricted mucosal T cells that are specific for ingested wheat gluten peptides, is in many ways ideal for this purpose. In addition, there is a need for alternative treatment to the gluten-free diet in this disorder. Here we have assessed the effects of atorvastatin on gluten-reactive T cells, dendritic cells and the coeliac mucosa by in vitro culture of biopsies. Atorvastatin inhibited gluten-induced proliferation and specific cytokine production of human intestinal gluten-reactive T cell clones and lines. Dendritic cells exposed to atorvastatin displayed a reduced expression of the costimulatory molecule CD83 upon maturation with lipopolysaccharide. Incubation of intestinal biopsy specimens with atorvastatin in vitro, however, did not influence gluten-induced cytokine release. In conclusion, atorvastatin has specific effects on isolated gluten-reactive T cells and dendritic cells, but does not shut down the gluten-induced production of proinflammatory cytokines in intestinal biopsies.
Collapse
Affiliation(s)
- M Ráki
- Institute of Immunology, University of Oslo, Rikshospitalet University Hospital, Norway.
| | | | | | | | | |
Collapse
|
226
|
Abstract
Celiac disease is a multigenetic complex inflammatory disorder with an autoimmune component, induced by gluten, a protein found in wheat. It is a unique human disease model to dissect the innate and adaptive immune mechanisms underlying T-cell-mediated tissue destruction and the development of T-cell lymphoma in conditions of chronic T-cell activation.
Collapse
Affiliation(s)
- Bana Jabri
- Department of Pathology, Medicine and Pediatrics, University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
227
|
Abstract
Celiac disease is manifested by an enteropathy caused by intolerance to gluten, a family of proteins found in wheat and other cereals. Following intestinal T-cell activation in predisposed individuals, different inflammatory mechanisms are triggered under the control of the cytokine balance including those with a pro-inflammatory Th1 pattern such as IFNgamma, TNFalpha, IL-15 and IL-18; and regulatory cytokines such as TGFbeta and IL-10. These cytokines, besides increasing the intensity of the activation and the number of immune cells within the intestinal mucosa, regulate the activity of epithelial growth factors and metalloproteinases, a group of molecules involved in the maintenance and turnover of the intestinal mucosa structure; in inflammatory conditions, they also induce the intestinal lesion responsible for malabsorption syndrome.
Collapse
Affiliation(s)
- Alberto J León
- Departamento de Pediatría e Inmunología, Instituto de Biología y Genética Molecular, Universidad de Valladolid, Valladolid, Spain
| | | | | |
Collapse
|
228
|
Spaenij-Dekking L, Kooy-Winkelaar Y, van Veelen P, Drijfhout JW, Jonker H, van Soest L, Smulders MJM, Bosch D, Gilissen LJWJ, Koning F. Natural variation in toxicity of wheat: potential for selection of nontoxic varieties for celiac disease patients. Gastroenterology 2005; 129:797-806. [PMID: 16143119 DOI: 10.1053/j.gastro.2005.06.017] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Accepted: 05/26/2005] [Indexed: 01/28/2023]
Abstract
BACKGROUND & AIMS Celiac disease (CD) is an intestinal disorder caused by T-cell responses to peptides derived from the gluten proteins present in wheat. Such peptides have been found both in the gliadin and glutenin proteins in gluten. The only cure for CD is a lifelong gluten-free diet. It is unknown, however, if all wheat varieties are equally harmful for patients. We investigated whether wheat varieties exist with a natural low number of T-cell-stimulatory epitopes. METHODS Gluten proteins present in public databases were analyzed for the presence of T-cell-stimulatory sequences. In addition, wheat accessions from diploid (AA, SS/BB, and DD genomes), tetraploid (AABB), and hexaploid (AABBDD) Triticum species were tested for the presence of T-cell-stimulatory epitopes in gliadins and glutenins by both T-cell and monoclonal antibody-based assays. RESULTS The database analysis readily identified gluten proteins that lack 1 or more of the known T-cell-stimulatory sequences. Moreover, both the T-cell- and antibody-based assays showed that a large variation exists in the amount of T-cell-stimulatory peptides present in the wheat accessions. CONCLUSIONS Sufficient genetic variation is present to endeavor the selection of wheat accessions that contain low amounts of T-cell-stimulatory sequences. Such materials may be used to select and breed wheat varieties suitable for consumption by CD patients, contributing to a well-balanced diet and an increase in their quality of life. Such varieties also may be useful for disease prevention in individuals at risk.
Collapse
Affiliation(s)
- Liesbeth Spaenij-Dekking
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Abstract
Coeliac disease is a common condition and its prevalence in UK is now thought to be approximately 1:100. It is being diagnosed and treated more frequently as awareness at the primary care level has increased. Coeliac disease is a complex disorder and is frequently associated with other disease processes. The management of these patients needs to take on a holistic approach, whilst the physician needs to be aware of the rare complications. This article gives an up-to-date review of the literature written on the pathogenesis of coeliac disease. We have attempted to paint a picture from beginning to end, whilst clarifying the grey areas in between. General epidemiological factors are reviewed before looking at genetic risk factors. We assess the sensitivity and specificity of the investigative modalities available for clinical use and comment on optimum management of these patients thereafter. The future of coeliac disease looks promising for patients with several novel therapies on the horizon. Whilst further work is still needed to breed out the toxic epitopes from wheat, novel therapies may come from other areas such as the work aimed at restoring normal tolerance to gluten.
Collapse
Affiliation(s)
- Paul J Ciclitira
- Department of Gastroenterology, GKT, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, United Kingdom.
| | | | | | | |
Collapse
|
230
|
Rueda B, Zhernakova A, López-Nevot MA, Martín J, Koeleman BPC. Association study of functional genetic variants of innate immunity related genes in celiac disease. BMC MEDICAL GENETICS 2005; 6:29. [PMID: 16078996 PMCID: PMC1190178 DOI: 10.1186/1471-2350-6-29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Accepted: 08/03/2005] [Indexed: 11/10/2022]
Abstract
Background Recent evidence suggest that the innate immune system is implicated in the early events of celiac disease (CD) pathogenesis. In this work for the first time we have assessed the relevance of different proinflammatory mediators typically related to innate immunity in CD predisposition. Methods We performed a familial study in which 105 celiac families characterized by the presence of an affected child with CD were genotyped for functional polymorphisms located at regulatory regions of IL-1α, IL-1β, IL-1RN, IL-18, RANTES and MCP-1 genes. Familial data was analysed with a transmission disequilibrium test (TDT) that revealed no statistically significant differences in the transmission pattern of the different genetic markers considered. Results The TDT analysis for IL-1α, IL-1β, IL-1RN, IL-18, and MCP-1 genes genetic variants did not reveal biased transmission to the affected offspring. Only a borderline association of RANTES promoter genetic variants with CD predisposition was observed. Conclusion Our results suggest that the analysed polymorphisms of IL-1α, IL-1β, IL-1RN, IL-18, RANTES and MCP-1 genes do not seem to play a major role in CD genetic predisposition in our population.
Collapse
Affiliation(s)
- B Rueda
- Instituto de Parasitología y Biomedicina "López-Neyra", CSIC, Granada, Spain
| | - A Zhernakova
- Complex Genetics Group, Department of Biomedical Genetics, University Medical Center, Utrecht, The Netherlands
| | - MA López-Nevot
- Servicio de Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - J Martín
- Instituto de Parasitología y Biomedicina "López-Neyra", CSIC, Granada, Spain
| | - BPC Koeleman
- Complex Genetics Group, Department of Biomedical Genetics, University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
231
|
Chakir H, Lefebvre DE, Wang H, Caraher E, Scott FW. Wheat protein-induced proinflammatory T helper 1 bias in mesenteric lymph nodes of young diabetes-prone rats. Diabetologia 2005; 48:1576-84. [PMID: 16003532 DOI: 10.1007/s00125-005-1842-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Accepted: 03/23/2005] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is the result of an inflammatory T helper 1 (Th1) lymphocyte-mediated beta cell destructive process. The majority of diabetes-prone BioBreeding (BBdp) rats fed wheat protein-based diets, such as NTP-2000, develop type 1 diabetes and display a mild coeliac-like enteropathy. Mesenteric lymph nodes (MLNs), which drain the gut, are the major inductive site where dietary antigens are recognised in the gut-associated lymphoid tissue (GALT). We hypothesised that this compartment could be a site of abnormal wheat protein-induced Th1 cell activation. METHODS MLN cells were isolated from BBdp and BB control (BBc) rats that were fed NTP-2000 or a hydrolysed casein (HC)-based diet at ages that pre-date classic insulitis. The inflammatory status, phenotype and proliferation of these cells in response to wheat protein were determined. RESULTS The expression ratio of T-bet : Gata3, master transcription factors for Th1 and Th2 cytokines, was increased in the MLN from NTP-2000-fed BBdp rats compared with that from BBc rats, mainly due to decreased Gata3 expression. CD3(+)CD4(+)IFN-gamma(+) T cells were more prevalent in the MLN of wheat-fed BBdp rats, but remained at control levels in BBdp rats fed a diabetes-retardant HC diet. BBdp MLN cells proliferated in response to wheat protein antigens in a specific, dose-dependent manner, and >93% of cells were CD3(+)CD4(+) T cells. This proliferation was associated with a low proportion of CD4(+)CD25(+) T cells and a high proportion of dendritic cells in the MLN of BBdp rats. CONCLUSIONS/INTERPRETATION Before insulitis is established, the MLNs of wheat-fed BBdp rats contain an unusually high proportion of Th1 cells that proliferate specifically in response to wheat protein antigens.
Collapse
Affiliation(s)
- H Chakir
- Molecular Medicine, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | | | | | | | | |
Collapse
|
232
|
Paajanen L, Vaarala O, Karttunen R, Tuure T, Korpela R, Kokkonen J. Increased IFN-gamma secretion from duodenal biopsy samples in delayed-type cow's milk allergy. Pediatr Allergy Immunol 2005; 16:439-44. [PMID: 16101938 DOI: 10.1111/j.1399-3038.2005.00312.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A delayed and local gastrointestinal hypersensitivity to cow's milk (CM) protein is difficult to diagnose and there are limited data on this disorder. The aim of this study was to investigate local intestinal cytokine secretion in the upper small intestine in children with delayed-type cow's milk allergy (CMA). Duodenal biopsy samples from 31 children with delayed CMA, 14 with celiac disease (CD), and 14 healthy controls were studied for the spontaneous release of IFN-gamma, TNF-alpha, IL-2, IL-4, IL-5, and IL-10, measured by cytometric bead array, and of TGF-beta and IL-6 measured by ELISA. The children with delayed CMA secreted more IFN-gamma than the controls (p = 0.006) and the children with CD (p = 0.006). The children with CD secreted more IL-6 compared to the controls (p = 0.008) and the children with delayed CMA (p = 0.002). The children with delayed CMA who had continuously been exposed to CM secreted less TGF-beta than the children with delayed CMA who avoided CM (p = 0.050), and showed a tendency towards lower secretion compared to the controls (p = 0.078). Secretions of TNF-alpha, IL-2, IL-4, IL-5, and IL-10 were low in general; however, the children with delayed CMA who did not avoid CM secreted more IL-4 and IL-10 than the controls (p = 0.016, 0.059). In conclusion, the children with delayed CMA showed up-regulation of IFN-gamma. Interestingly, TGF-beta secretion was up-regulated in those children with delayed CMA who avoided CM suggesting recovery of regulation mechanisms.
Collapse
Affiliation(s)
- Laura Paajanen
- Foundation for Nutrition Research, PO Box 30, FIN-00039 Valio, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
233
|
Rueda B, Koeleman BPC, López-Nevot MA, Ortega E, Maldonado J, López M, Polanco I, Martín J. Poly (ADP-ribose) polymerase-1 haplotypes are associated with coeliac disease. Int J Immunogenet 2005; 32:245-8. [PMID: 16026592 DOI: 10.1111/j.1744-313x.2005.00521.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In coeliac disease (CD) there is an inflammatory status of the intestinal mucosa because of a high expression of proinflammatory mediators. The nuclear protein poly (ADP-ribose) polymerase-1 (PARP-1) has been implicated in the initial inflammatory response by modulating transcription of inflammation-related genes. The aim of this work was to investigate the role of PARP-1 gene promoter region haplotypes in relation to coeliac disease susceptibility. We analysed a coeliac population consisting of a case-control panel with 120 CD patients and 311 healthy blood donors. A CA microsatellite, as haplotype-defining variant of the whole PARP-1 promoter, was typed using a polymerase chain reaction (PCR) method combined with fluorescence technology. We considered two promoter haplotypes: A defined by short CA alleles (83-87 bp) and B defined by long CA alleles (89-101 bp). Haplotype A was significantly increased within the coeliac patients group (P = 0.007 OR 1.6 95%CI 1.12-2.35). Additionally, we observed a significant dose effect, showing homozygous individuals for haplotype A higher risk for CD susceptibility (P = 0.007, OR 1.79 95%CI 1.14-2.82). Our results provide the first evidence that PARP-1 haplotypes are related with coeliac disease susceptibility.
Collapse
Affiliation(s)
- B Rueda
- Instituto de Parasitología y Biomedicina López-Neyra (CSIC), Parque Tecnológico de Ciencias de la Salud, Avda. del Conocimiento, s/n, 18100 Armilla, Granada, Spain
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Ebert EC. IL-15 converts human intestinal intraepithelial lymphocytes to CD94 producers of IFN-gamma and IL-10, the latter promoting Fas ligand-mediated cytotoxicity. Immunology 2005; 115:118-26. [PMID: 15819704 PMCID: PMC1782126 DOI: 10.1111/j.1365-2567.2005.02132.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs), T-cell receptor alphabeta(+) CD8(+) T cells located between epithelial cells, are thought to contribute to Fas ligand (FL)-mediated epithelial cell death in coeliac disease, a condition characterized by excess interleukin-15 (IL-15). This study evaluates the effects of prolonged IL-15 stimulation on IELs. Human IELs were obtained from jejunal mucosa from gastric bypass operations for morbid obesity and cultured for 3 or 10 days with IL-15. As the culture progressed, an increasing number of IELs became CD94(+) and produced massive quantities of interferon-gamma (IFN-gamma) and IL-10. There was a steady rate of transcription with no feedback regulation. Few chronically activated IELs produced IL-2, IL-4, or tumour necrosis factor-alpha (TauNuF-alpha). To determine whether the accumulation of IL-10 affected IEL functions, endogenous IL-10 was neutralized by antibody during culture with IL-15. This manipulation reduced expression of CD94, NKG2D, and FL as well as FL-mediated killing of Jurkat cells by IELs. It did not affect perforin or TNF-alpha expression or the associated cytotoxic activities. This study shows that IL-15 induces the development of CD94(+) IELs containing IFN-gamma and IL-10, and that endogenous IL-10 promotes FL-mediated cytotoxicity.
Collapse
Affiliation(s)
- Ellen C Ebert
- UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA.
| |
Collapse
|
235
|
Londei M, Ciacci C, Ricciardelli I, Vacca L, Quaratino S, Maiuri L. Gliadin as a stimulator of innate responses in celiac disease. Mol Immunol 2005; 42:913-8. [PMID: 15829281 DOI: 10.1016/j.molimm.2004.12.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In celiac disease (CD) we have the prototype of an immune mediated response dominated by the activation of the adaptive immune system and in particular of CD4+ HLA class II restricted T cells. Various seminal studies have established the precise mechanism of how antigen (prolamine) specific activation of CD4+ mucosal T cells occurs. Thus, CD is a condition in which T cells and their activation is the essential hinge in the pathogenic process. These functional studies have provided the explanation for the genetic association between CD and certain HLA alleles (HLA DQ2 and DQ8). These genetic, molecular and functional studies have permitted the clarification of a powerful Th1 dominated pro-inflammatory response that characterises the small intestine of active CD patients. Despite this unassailable set of information and reports there are some intriguing points that have been raised by a series of studies which have indicated that CD is not only defined by an aberrant prolamine-induced activation of the adaptive immune system. New evidence and re-assessments of old studies, point to a more complex pathogenic cascade, which may help to unravel some of the residual obscure points of CD pathogenesis. Here, we outline the current concepts that indicate a direct involvement of the adaptive immune system and we discuss all the evidence supporting a direct activation of the innate immune system by fragments of prolamines, which are not recognized T cell epitopes and how they could influence CD. The gliadin-induced activation of the 'innate' immune system might also have a significant role in the induction and persistence of many CD complications and most definitively for the most aggressive one, namely mucosal T cell lymphomas. We further suggest a novel way to harness the unwanted immune response to toxic prolamine, and thus indicate new potential therapeutic strategies to treat or at least control CD.
Collapse
Affiliation(s)
- Marco Londei
- Marco Londei Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.
| | | | | | | | | | | |
Collapse
|
236
|
Abstract
Celiac disease is a chronic enteropathy caused by intolerance to gluten. The true prevalence of this condition is much greater than previously recognized, with increasing numbers of silent cases being diagnosed. Population-based studies, using serologic screening, have indicated that the prevalence of celiac disease in Caucasian populations is .5%-1%. The pattern of incidence is changing, with a greater proportion of cases diagnosed later in adulthood. The pathologic lesion is characterized by a flattened small intestinal mucosa with a lymphocytic infiltrate, crypt hyperplasia, and villous atrophy. Absorptive function may be impaired and patients can experience gastrointestinal symptoms and malabsorption leading to development of anemia, osteoporosis, or other complications. Untreated celiac disease is associated with significant morbidity and increased mortality, largely owing to the development of enteropathy-associated intestinal lymphoma. The pathologic changes and symptoms resolve when gluten is excluded from the diet for a sustained period.
Collapse
Affiliation(s)
- David H Dewar
- Department of Gastroenterology, Guy's, King's and St Thomas' Medical School, The Rayne Institute, St Thomas' Hospital, London, United Kingdom.
| | | |
Collapse
|
237
|
Haddeland U, Sletten GB, Brandtzaeg P, Nakstad B. Impaired interleukin (IL)-4-associated generation of CCR4-expressing T cells in neonates with hereditary allergy risk. Clin Exp Immunol 2005; 139:314-22. [PMID: 15654830 PMCID: PMC1809286 DOI: 10.1111/j.1365-2249.2005.02706.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reduced microbial exposure in early life may contribute to the increase of atopic diseases in 'westernized' societies but the underlying mechanisms remain elusive. The objective of this study was to examine how exposure to bacterial lipopolysaccharide (LPS) during early antigen encounter might influence the maturation of neonatal lymphoid cells, and to define possible differences in this respect between neonates with high risk of allergy due to a family history (FH(+)) and controls with no apparent hereditary risk (FH(-)). Cord blood mononuclear cells from the FH(+) or FH(-) group were stimulated with pure LPS or beta-lactoglobulin (beta-LG) in the presence of LPS. T cell expression of chemokine receptors CCR4 and CXCR3 was determined by flow cytometry and reverse transcription-polymerase chain reaction (RT-PCR). Cellular expression of interleukin (IL)-4 was analysed by quantitative RT-PCR, whereas interferon (IFN)-gamma was analysed by both quantitative RT-PCR and immunoassay. Stimulation with LPS, or beta-LG together with LPS, induced up-regulation of CCR4 (P < 0.05) and CXCR3 (P < 0.05). For CCR4, such up-regulation was related to the level of IL-4 produced by the same T cells (r(S) = 0.49, P = 0.03), while CXCR3 expression was negatively correlated with the IL-4 levels (r(S) = -0.56, P = 0.02). Compared with the FH(-) group, the FH(+) group showed a significantly lower capacity for generation of CCR4(+) T cells (mean percentage of total T cells: FH(+), 2.42%versus FH(-), 5.74%; P < 0.01), whereas induction of CXCR3 and IFN-gamma did not differ significantly between the two groups. When the immune system in early life encounters antigen together with LPS, the T cell potential for compartmentalized interaction with other immune cells might be increased by elevated CCR4- and CXCR3-expression levels. In neonates at hereditary allergy risk, this putative homeostatic mechanism could theoretically be jeopardized due to decreased up-regulation of CCR4. Conversely, Th1 responses to antigen in the presence of LPS did not appear to be reduced compared with controls.
Collapse
Affiliation(s)
- U Haddeland
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, Rikshospitalet University Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
238
|
Lettesjö H, Hansson T, Bergqvist A, Grönlund J, Dannaeus A. Enhanced interleukin-18 levels in the peripheral blood of children with coeliac disease. Clin Exp Immunol 2005; 139:138-43. [PMID: 15606624 PMCID: PMC1809275 DOI: 10.1111/j.1365-2249.2005.02661.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Coeliac disease (CoD) is a small intestinal disorder characterized by villous atrophy, crypt cell hyperplasia and an increased production of T helper cell type 1 (Th1) cytokines. Interleukin (IL)-18 is a pro-inflammatory cytokine that has a crucial role in maintaining the Th1 response. In this study, the serum levels of IL-18 were measured in children with CoD or other gastrointestinal diseases in order to evaluate the possibility of using IL-18 as a disease activity marker. IL-18 levels were higher in samples from CoD patients [median 443 pg/ml (148-885)] compared to healthy controls [median 205 pg/ml (11-379)], P <0.05. In contrast, the levels of IL-18 were not enhanced significantly in the serum from patients with inflammatory bowel disease (IBD) [median 324 pg/ml (207-546)] or in the disease control group [median 303 pg/ml (2-689)]. In CoD patients, after 2 weeks of gluten challenge (GC), serum IL-18 was unchanged [median 268 pg/ml (59-458)] compared to patients on a gluten-free diet [median 220 pg/ml (53-600)], while IL-18 was increased after 12 weeks of GC [median 551 pg/ml (94-952)], P <0.01. The IL-18 levels correlated with IgA anti-transglutaminase antibody levels (rs=0.59, P=0.016) in serum from untreated CoD patients, and IL-18 also followed the degree of small intestinal villous atrophy in 12 out of 19 CoD patients. Our results support the view that serum IL-18 concentrations in children with CoD follow disease activity, suggesting a role for IL-18 in the induction of an inflammatory Th1-response after gluten exposure.
Collapse
Affiliation(s)
- H Lettesjö
- Department of Gastrointestinal Research, Pharmacia Diagnostics, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
239
|
León F, Sánchez L, Camarero C, Roy G. Cytokine production by intestinal intraepithelial lymphocyte subsets in celiac disease. Dig Dis Sci 2005; 50:593-600. [PMID: 15810648 DOI: 10.1007/s10620-005-2480-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the earliest signs of mucosal immune activation in celiac disease (CD) is an increase in the intraepithelial lymphocyte (IEL) count in the small intestinal epithelium. Though most of those IELs express T cell receptor (TcR)-alphabeta chains, CD is characterized by an increase in TcR-gammadelta+ IELs and by the loss of CD3- IELs. There is currently little evidence that these changes in IEL subset distribution are of relevance in the pathogenesis of CD. We aimed to determine the pattern of cytokine production by IEL subsets isolated from duodenal biopsy specimens from control subjects and CD patients at different stages of the disease. We quantified the capacity of IEL subsets to produce IFN-gamma, TNF-alpha, IL-2, IL-4, and IL-10 by intracellular staining by flow cytometry. All IEL subsets studied displayed a type I cytokine profile in both CD and control subjects, with TcR-alphabeta+ IELs being the main IFN-y producers. Untreated CD exhibited a trend toward a superior accumulation of IFN-gamma per cell but a reduced proportion of INF-gamma+ cells in vitro in association with a significantly increased apoptotic rate of IELs. IL-4 was almost undetectable in all cases and IL-10 showed a tendency to increase in treated and "silent" celiac patients. IEL subsets have a similar Th1 profile in controls and CD patients, and the superior in vitro apoptosis of IELs from CD patients may reflect their superior in vivo activation. The induction of IL-10-dependent regulatory Tr1 responses may be of potential clinical significance in this disease and merits further investigation.
Collapse
Affiliation(s)
- Francisco León
- Departments of Immunology and Paediatrics, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | |
Collapse
|
240
|
Ciccocioppo R, Di Sabatino A, Bauer M, Della Riccia DN, Bizzini F, Biagi F, Cifone MG, Corazza GR, Schuppan D. Matrix metalloproteinase pattern in celiac duodenal mucosa. J Transl Med 2005; 85:397-407. [PMID: 15608660 DOI: 10.1038/labinvest.3700225] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of endopeptidases playing a key role in tissue remodelling in both physiological and pathological conditions. Since little information is available about their role in celiac disease (CD), our aims were to quantify their expression/activity and to investigate their relation to proinflammatory cytokines in this condition. Duodenal biopsies from untreated, treated celiac patients and controls were used to quantify the expression of MMP-1, MMP-2, MMP-3, MMP-9, MMP-12, MMP-14, their inhibitor TIMP-1, IFN-gamma and TNF-alpha by using real-time reverse transcription-polymerase chain reaction and the gelatin/casein/elastin activities by gel zymography, and to isolate lamina propria mononuclear cells (LPMCs). These cells and myofibroblasts isolated from jejunal specimens were incubated in the absence or presence of IFN-gamma and TNF-alpha. MMP-1 and MMP-12 mRNA levels were significantly increased in active CD compared to treated (P<0.01 and P<0.0005, respectively) and normal mucosa (P<0.01 and P<0.0005, respectively), and this was paralleled by an upregulation of caseinolytic and elastolytic activities. Furthermore, MMP-12 levels significantly (P<0.05) correlated with those of IFN-gamma and the degree of villous flattening. MMP-2 turned out to be significantly (P<0.05) reduced in untreated and treated celiacs compared to controls. In active CD, transcripts of TIMP-1 were higher than in treated and controls (P<0.005 and P<0.05, respectively), such as those of IFN-gamma (P<0.05), whereas TNF-alpha levels were suppressed (P=0.0001). In physiological condition, myofibroblasts represent the main source of MMP-2, whereas LPMCs produce almost all MMPs only after cytokine stimulation. Conversely, cells isolated from active patients constitutively express MMPs without any increase after cytokine stimulation, while those from treated patients are in a resting condition. In conclusion, our results show the presence of a peculiar MMP pattern in active CD strongly dominated by MMP-12, correlating either with IFN-gamma or the degree of mucosal damage.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, IRCCS Policlinico San Matteo, University of Pavia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
241
|
|
242
|
Peláez-Luna M, Schmulson M, Robles-Díaz G. Intestinal involvement is not sufficient to explain hypertransaminasemia in celiac disease? Med Hypotheses 2005; 65:937-941. [PMID: 16023789 DOI: 10.1016/j.mehy.2005.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 05/05/2005] [Accepted: 05/09/2005] [Indexed: 01/10/2023]
Abstract
Chronic unexplained hypertransaminasemia is an isolated clinical manifestation of celiac disease (CD) and lacks of a clear physiopathological explanation. Since CD and tropical sprue (TS) have similar intestinal functional and histological pattern of injury and that an increased inflammatory response has been reported to occur in patients with irritable bowel syndrome (IBS), liver involvement might be expected to occur either in TS or IBS. However, according to author's prior observations, the frequency of hypertransaminasemia is significantly higher in CD than in TS and IBS-diarrhea predominant patients (IBS-D). Thus, based on current knowledge, intestinal mucosal damage, increased intestinal permeability and/or an active intestinal inflammatory response do not completely explain liver damage in CD. We hypothesize that other factors, unique to CD not present in TS or IBS-D, like gluten toxicity and the presence of tissular transglutaminase (tTG) an auto-antigen with pro-inflammatory and remodeling properties, act in addition to intestinal mucosal injury and account to hypertransaminasemia in CD. Further research focusing on the mechanisms of gluten and tTG hepatic toxicity, and/or the characterization of the expression, secretion and enteral-hepatic transport of certain pro-inflammatory cytokines is needed, to understand the possible links between intestinal and liver disorders seen in CD.
Collapse
Affiliation(s)
- Mario Peláez-Luna
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Colonial Sección XVI, Tlalpan 14000, México City, Mexico
| | | | | |
Collapse
|
243
|
Salvati VM, Mazzarella G, Gianfrani C, Levings MK, Stefanile R, De Giulio B, Iaquinto G, Giardullo N, Auricchio S, Roncarolo MG, Troncone R. Recombinant human interleukin 10 suppresses gliadin dependent T cell activation in ex vivo cultured coeliac intestinal mucosa. Gut 2005. [PMID: 15591503 DOI: 10.1136/gut.2003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
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.
Collapse
Affiliation(s)
- V M Salvati
- Department of Paediatrics, University Federico II, Via Pansini, No 5, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Woolley N, Mustalahti K, Mäki M, Partanen J. Cytokine Gene Polymorphisms and Genetic Association with Coeliac Disease in the Finnish Population. Scand J Immunol 2005; 61:51-6. [PMID: 15644122 DOI: 10.1111/j.0300-9475.2005.01525.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Coeliac disease (CD) is an intestinal disorder caused by intolerance to dietary gluten in susceptible individuals. The HLA-DQ genes are major risk factors for CD, but other genes also play an important role in the disease susceptibility. Immune-mediated mechanisms are known to underlie the pathogenesis of CD. We studied single-nucleotide polymorphisms in transforming growth factor (TGF)-beta1, interleukin (IL)-10, IL-6, interferon (IFN)-gamma and tumour necrosis factor (TNF)-alpha genes in the Finnish population using family-based association approach. In addition, we genotyped a trinucleotide repeat polymorphism in the major histocompatibility complex (MHC) class I chain-related protein A (MICA) gene, located in the human leucocyte antigen (HLA) region in the vicinity of TNF-alpha. To control the effect of linkage disequilibrium between HLA-DQ genes and MICA and TNF-alpha, an HLA-stratified association analysis was performed. We did not find evidence of association between TGF-beta1, IL-10, IL-6 and IFN-gamma polymorphisms and CD susceptibility. No association was found for any of the MICA alleles independently of DQ genes, whereas TNF-alpha-308 A allele was slightly overrepresented on chromosomes carried by CD patients compared with control chromosomes, indicating that either TNF-alpha, or another gene in linkage disequilibrium with it, could confer increased susceptibility to CD. This result supports the earlier findings that the HLA region harbours a novel susceptibility factor in addition to HLA-DQ.
Collapse
Affiliation(s)
- N Woolley
- Research Laboratory and Department of Tissue Typing, Finnish Red Cross Blood Service, Helsinki, Finland.
| | | | | | | |
Collapse
|
245
|
Yan F, John SK, Wilson G, Jones DS, Washington MK, Polk DB. Kinase suppressor of Ras-1 protects intestinal epithelium from cytokine-mediated apoptosis during inflammation. J Clin Invest 2004; 114:1272-80. [PMID: 15520859 PMCID: PMC524224 DOI: 10.1172/jci21022] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Accepted: 08/10/2004] [Indexed: 01/22/2023] Open
Abstract
TNF plays a pathogenic role in inflammatory bowel diseases (IBDs), which are characterized by altered cytokine production and increased intestinal epithelial cell apoptosis. In vitro studies suggest that kinase suppressor of Ras-1 (KSR1) is an essential regulatory kinase for TNF-stimulated survival pathways in intestinal epithelial cell lines. Here we use a KSR1-deficient mouse model to study the role of KSR1 in regulating intestinal cell fate during cytokine-mediated inflammation. We show that KSR1 and its target signaling pathways are activated in inflamed colon mucosa. Loss of KSR1 increases susceptibility to chronic colitis and TNF-induced apoptosis in the intestinal epithelial cell. Furthermore, disruption of KSR1 expression enhances TNF-induced apoptosis in mouse colon epithelial cells and is associated with a failure to activate antiapoptotic signals including Raf-1/MEK/ERK, NF-kappaB, and Akt/protein kinase B. These effects are reversed by WT, but not kinase-inactive, KSR1. We conclude that KSR1 has an essential protective role in the intestinal epithelial cell during inflammation through activation of cell survival pathways.
Collapse
Affiliation(s)
- Fang Yan
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2576, USA
| | | | | | | | | | | |
Collapse
|
246
|
Skovbjerg H, Hansen GH, Niels-Christiansen LL, Anthonsen D, Ascher H, Midhagen G, Hallert C, Norén O, Sjöström H. Intestinal tissue transglutaminase in coeliac disease of children and adults: ultrastructural localization and variation in expression. Scand J Gastroenterol 2004; 39:1219-27. [PMID: 15742999 DOI: 10.1080/00365520410003597] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Tissue transglutaminase is the main antigen for the anti-endomysial antibodies used for diagnosis of coeliac disease and can with some specificity in vitro deamidate gliadins generating potent epitopes. The intestinal levels and the ultrastructural localization of tissue transglutaminase in normal and affected persons were investigated to provide further information on its role in this disease. Intestinal biopsies were taken from normal and coeliac children and adults. METHODS The level of transglutaminase was analysed by means of a quantitative enzymatic assay and its ultrastructural localization by immunogold electronmicroscopy using a monoclonal antibody against tissue transglutaminase. RESULTS In relation to normal individuals, the enzymatic activity of tissue transglutaminase in adult coeliac patients was increased. The enzyme was found in the enterocytes and in increased amount just beneath the enterocytes, where cytosolic and nuclear labelling of distinct elongated cells was seen in addition to extracellular labelling close to collagen fibrils. In children, the enzymatic activity and the immunogold labelling could not be shown to be related to disease. In all cases the enzyme activity was EDTA-sensitive. CONCLUSIONS The increased amount of tissue transglutaminase activity in coeliac adults was shown to be due to the appearance of the enzyme in enterocytes and increased expression in the lamina propria. No evidence was found to support the idea of a changed localization or changed amounts as primary elements in coeliac disease pathogenesis, nor for the involvement of non-calcium dependent microbial transglutaminases.
Collapse
Affiliation(s)
- H Skovbjerg
- Biochemistry Laboratory C, Dept of Medical Biochemistry and Genetics, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Cao W, Cheng L, Behar J, Fiocchi C, Biancani P, Harnett KM. Proinflammatory cytokines alter/reduce esophageal circular muscle contraction in experimental cat esophagitis. Am J Physiol Gastrointest Liver Physiol 2004; 287:G1131-9. [PMID: 15271650 DOI: 10.1152/ajpgi.00216.2004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cholinergic mechanisms are largely responsible for esophageal contraction in response to swallowing or to in vitro electrical field stimulation (EFS). After induction of experimental esophagitis by repeated acid perfusion, the responses to swallowing and to EFS were significantly reduced but contraction in response to ACh was not affected, suggesting that cholinergic mechanisms are damaged by acid perfusion but that myogenic mechanisms are not. Measurements of ACh release in response to EFS confirmed that release of ACh was reduced in esophagitis compared with normal controls. To examine factors contributing to this neuropathy, normal esophageal strips were incubated for 1-2 h with the proinflammatory cytokines IL-1beta (100 U/ml), IL-6 (1 ng/ml), or TNF-alpha (1 ng/ml). IL-1beta and IL-6 levels, measured by Western blot analysis, increased in esophagitis compared with normal circular muscle. IL-1beta and IL-6 reduced contraction in response to EFS (2-10 Hz, 0.2 ms) but did not affect ACh-induced contraction, suggesting that these cytokines inhibit ACh release without affecting myogenic contractile mechanisms. EFS-induced ACh release was significantly reduced in normal esophageal strips by incubation in IL-1beta or IL-6, suggesting that they may contribute to the contractility changes. TNF-alpha at 1 ng/ml, however, did not affect the response to ACh or to electrical stimulation but inhibited both at higher concentrations. TNF-alpha levels were low in normal muscle and did not increase with esophagitis. The data suggest that the proinflammatory cytokines IL-1beta and IL-6 contribute to reduced esophageal contraction by inhibiting release of ACh from myenteric neurons.
Collapse
Affiliation(s)
- Weibiao Cao
- Department of Medicine, Rhode Island Hospital and Brown University, 593 Eddy St., Providence, RI 02903, USA
| | | | | | | | | | | |
Collapse
|
248
|
Yan F, John SK, Wilson G, Jones DS, Washington MK, Polk DB. Kinase suppressor of Ras-1 protects intestinal epithelium from cytokine-mediated apoptosis during inflammation. J Clin Invest 2004. [DOI: 10.1172/jci200421022] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
249
|
Durante-Mangoni E, Iardino P, Resse M, Cesaro G, Sica A, Farzati B, Ruggiero G, Adinolfi LE. Silent celiac disease in chronic hepatitis C: impact of interferon treatment on the disease onset and clinical outcome. J Clin Gastroenterol 2004; 38:901-5. [PMID: 15492610 DOI: 10.1097/00004836-200411000-00014] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GOALS To assess the impact of interferon treatment on celiac disease onset in hepatitis C patients and to clarify its clinical relevance and outcome. BACKGROUND Hepatitis C is associated with autoimmunity, which can be exacerbated by interferon treatment. Cases of celiac disease activation during interferon treatment have been reported. STUDY Retrospective evaluation of 534 hepatitis C patients with or without symptoms compatible with celiac disease onset during interferon treatment and 225 controls. Anti-transglutaminase antibodies were assayed. HLA-DQA1 and -B1 loci were typed. Upper gastrointestinal endoscopy was applied to confirm the diagnosis in antibody-positive patients. RESULTS Anti-transglutaminase antibodies were detected before treatment in 1.3% of hepatitis C patients and in 0.4% of controls (not significant). Eighty-six percent of patients with anti-transglutaminase antibodies showed activation of celiac disease while on interferon. Symptoms ranged from mild to severe, and interferon had to be discontinued in 2 of 7 (29%) patients. Symptoms disappeared in 6 of 7 patients after interferon withdrawal. Onset of symptoms compatible with celiac disease during interferon therapy was significantly associated with the presence of anti-transglutaminase antibodies (OR 53). CONCLUSIONS In hepatitis C patients, the activation of silent celiac disease during interferon treatment is almost universal and should be suspected, but it uncommonly requires interferon treatment discontinuation. Symptoms subside after interferon withdrawal.
Collapse
Affiliation(s)
- Emanuele Durante-Mangoni
- Divisions of Internal Medicine and Hepatology, Second University of Naples Medical School, Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|
250
|
Jelínková L, Tucková L, Cinová J, Flegelová Z, Tlaskalová-Hogenová H. Gliadin stimulates human monocytes to production of IL-8 and TNF-alpha through a mechanism involving NF-kappaB. FEBS Lett 2004; 571:81-5. [PMID: 15280021 DOI: 10.1016/j.febslet.2004.06.057] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Revised: 04/01/2004] [Accepted: 06/15/2004] [Indexed: 01/28/2023]
Abstract
Wheat gliadin is the triggering agent in coeliac disease. In this study, we documented that proteolytic fragments of gliadin, in contrast to other food antigens, induced interleukin (IL)-8 and tumour necrosis factor-alpha (TNF-alpha) production and significantly increased interferon (IFN)-gamma-induced cytokine secretion in human monocytic line THP-1 cells. Stimulation with gliadin resulted in elevated phosphorylation of the IkappaBalpha molecule and increased NF-kappaB/DNA binding activity that was inhibited by sulfasalazine, l-1-tosylamido-2-phenylethyl chloromethyl ketone and pyrrolidine dithiocarbamate (PDTC). The activation pathway was shown to be independent of the CD14 molecule. Less mature U-937 monocytes responded to gliadin stimulation by low IL-8 secretion, TNF-alpha production was not detectable. We propose that gliadin-induced activation of monocytes/macrophages can participate in mechanisms leading to the impairment of intestinal mucosa in coeliac patients.
Collapse
Affiliation(s)
- Lenka Jelínková
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídenská 1083, 142 20 Prague, Czech Republic.
| | | | | | | | | |
Collapse
|