51
|
Mowat AM. To respond or not to respond - a personal perspective of intestinal tolerance. Nat Rev Immunol 2019; 18:405-415. [PMID: 29491358 DOI: 10.1038/s41577-018-0002-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
For many years, the intestine was one of the poor relations of the immunology world, being a realm inhabited mostly by specialists and those interested in unusual phenomena. However, this has changed dramatically in recent years with the realization of how important the microbiota is in shaping immune function throughout the body, and almost every major immunology institution now includes the intestine as an area of interest. One of the most important aspects of the intestinal immune system is how it discriminates carefully between harmless and harmful antigens, in particular, its ability to generate active tolerance to materials such as commensal bacteria and food proteins. This phenomenon has been recognized for more than 100 years, and it is essential for preventing inflammatory disease in the intestine, but its basis remains enigmatic. Here, I discuss the progress that has been made in understanding oral tolerance during my 40 years in the field and highlight the topics that will be the focus of future research.
Collapse
Affiliation(s)
- Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
52
|
Rostami-Nejad M, vafaee R, Ehsani-Ardakani MJ, Aghamohammadi N, Keramatinia A, Abdi S, Moravvej H. The Screening of Critical Related Genes in Celiac Disease Based on Intraepithelial Lymphocytes Investigation: A Bioinformatics Analysis. Galen Med J 2019; 8:e1407. [PMID: 34466507 PMCID: PMC8343984 DOI: 10.31661/gmj.v8i0.1407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 11/16/2022] Open
Abstract
Background Celiac disease (CD) is an immunological intestinal disorder, which is characterized by response to gluten. In addition to the environmental factors and dysbiosis of the gut microbiota, genetic susceptibility has an important role in the pathogenesis of this multifactorial disorder. Therefore, this study aims to present the crucial involved genes in CD pathogenesis. Materials and Methods In this bioinformatics analysis study, significant differentially expressed genes of intraepithelial lymphocytes (IELs) samples of celiac patients versus normal patients from Gene Expression Omnibus (GEO) database were screened via the protein-protein interaction (PPI) network. The critical nodes based on degree values, betweenness centrality, and fold changes were determined and enriched by ClueGO to find relative biological terms. Results According to the network analysis, five central nodes including IL2, PIK3CA, PRDM10, AKT1, and SRC and eight significant differentially expressed genes (DEGs) were determined as the critical genes related to CD. Also, CD4+, CD25+, alpha-beta regulatory T cell differentiation are identified as prominent biological terms in the celiac disease patients. Conclusion There is a possible biomarker panel related to CD that can be used as a therapeutic or diagnostic tool to manage the disease.
Collapse
Affiliation(s)
- Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza vafaee
- Proteomics Research Center, Faculty of Paramedical Sciences, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Ehsani-Ardakani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nika Aghamohammadi
- Department of Dental Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Saeed Abdi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Correspondence to: Saeed Abdi, Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran Telephone Number: 02122432525 Email Address:
| | - Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
53
|
Vaquero L, Bernardo D, León F, Rodríguez-Martín L, Alvarez-Cuenllas B, Vivas S. Challenges to drug discovery for celiac disease and approaches to overcome them. Expert Opin Drug Discov 2019; 14:957-968. [DOI: 10.1080/17460441.2019.1642321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Luis Vaquero
- Gastroenterology Unit, University Hospital of León, León, Spain
| | - David Bernardo
- Mucosal Immunology lab, IBGM (University of Valladolid-CSIC), Valladolid, Spain
- Gut Immunology Research Lab, Instituto de Investigación Sanitaria Princesa (IIS-IP) & Centro de Investigación Biomédica en Red de Enfermdades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | | | - Laura Rodríguez-Martín
- Gastroenterology Unit, University Hospital of León, León, Spain
- Institute of Biomedicina (IBIOMED), University of León, León, Spain
| | | | - Santiago Vivas
- Gastroenterology Unit, University Hospital of León, León, Spain
- Institute of Biomedicina (IBIOMED), University of León, León, Spain
| |
Collapse
|
54
|
Coeliac Disease and Mast Cells. Int J Mol Sci 2019; 20:ijms20143400. [PMID: 31373285 PMCID: PMC6678566 DOI: 10.3390/ijms20143400] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023] Open
Abstract
Over the last decades, there has been an impressive progress in our understanding of coeliac disease pathogenesis and it has become clear that the disorder is the final result of complex interactions of environmental, genetic, and immunological factors. Coeliac disease is now considered a prototype of T-cell-mediated disease characterized by loss of tolerance to dietary gluten and the targeted killing of enterocytes by T-cell receptor αβ intraepithelial lymphocytes. Accumulating evidence, however, indicates that the induction of a gluten-specific T helper-1 response must be preceded by the activation of the innate immune system. Mast cells are key players of the innate immune response and contribute to the pathogenesis of a multitude of diseases. Here, we review the results of studies aimed at investigating the role of mast cells in the pathogenesis of coeliac disease, showing that these cells increase in number during the progression of the disease and contribute to define a pro-inflammatory microenvironment.
Collapse
|
55
|
Lerner A, Ramesh A, Matthias T. The Revival of the Battle between David and Goliath in the Enteric Viruses and Microbiota Struggle: Potential Implication for Celiac Disease. Microorganisms 2019; 7:173. [PMID: 31207872 PMCID: PMC6616392 DOI: 10.3390/microorganisms7060173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023] Open
Abstract
The human gut is inhabited by overcrowded prokaryotic communities, a major component of which is the virome, comprised of viruses, bacteriophages, archaea, eukaryotes and bacteria. The virome is required for luminal homeostasis and, by their lytic or synergic capacities, they can regulate the microbial community structure and activity. Dysbiosis is associated with numerous chronic human diseases. Since the virome can impact microbial genetics and behavior, understanding its biology, composition, cellular cycle, regulation, mode of action and potential beneficial or hostile activities can change the present paradigm of the cross-talks in the luminal gut compartment. Celiac disease is a frequent autoimmune disease in which viruses can play a role in disease development. Based on the current knowledge on the enteric virome, in relation to celiac disease pathophysiological evolvement, the current review summarizes the potential interphases between the two. Exploring and understanding the role of the enteric virome in gluten-dependent enteropathy might bring new therapeutic strategies to change the luminal eco-event for the patient's benefit.
Collapse
Affiliation(s)
- Aaron Lerner
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| | - Ajay Ramesh
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| | - Torsten Matthias
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| |
Collapse
|
56
|
Lerner A, Ramesh A, Matthias T. The Revival of the Battle between David and Goliath in the Enteric Viruses and Microbiota Struggle: Potential Implication for Celiac Disease. Microorganisms 2019; 7:173. [PMID: 31207872 DOI: 10.3390/microorganisms7060173.pmid:31207872;pmcid:pmc6616392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 05/12/2023] Open
Abstract
The human gut is inhabited by overcrowded prokaryotic communities, a major component of which is the virome, comprised of viruses, bacteriophages, archaea, eukaryotes and bacteria. The virome is required for luminal homeostasis and, by their lytic or synergic capacities, they can regulate the microbial community structure and activity. Dysbiosis is associated with numerous chronic human diseases. Since the virome can impact microbial genetics and behavior, understanding its biology, composition, cellular cycle, regulation, mode of action and potential beneficial or hostile activities can change the present paradigm of the cross-talks in the luminal gut compartment. Celiac disease is a frequent autoimmune disease in which viruses can play a role in disease development. Based on the current knowledge on the enteric virome, in relation to celiac disease pathophysiological evolvement, the current review summarizes the potential interphases between the two. Exploring and understanding the role of the enteric virome in gluten-dependent enteropathy might bring new therapeutic strategies to change the luminal eco-event for the patient's benefit.
Collapse
Affiliation(s)
- Aaron Lerner
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| | - Ajay Ramesh
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| | - Torsten Matthias
- AESKU.KIPP Institute, Mikroforum Ring 2, 55234 Wendelsheim, Germany.
| |
Collapse
|
57
|
Grover J, Chhuneja P, Midha V, Ghia JE, Deka D, Mukhopadhyay CS, Sood N, Mahajan R, Singh A, Verma R, Bansal E, Sood A. Variable Immunogenic Potential of Wheat: Prospective for Selection of Innocuous Varieties for Celiac Disease Patients via in vitro Approach. Front Immunol 2019; 10:84. [PMID: 30804930 PMCID: PMC6371638 DOI: 10.3389/fimmu.2019.00084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 01/11/2019] [Indexed: 01/26/2023] Open
Abstract
Celiac Disease (CD) is a multifactorial, autoimmune enteropathy activated by cereal proteins in genetically predisposed individuals carrying HLA DQ2/8 genes. A heterogenous gene combination of the cereal prolamins is documented in different wheat genotypes, which is suggestive of their variable immunogenic potential. In the current study, four wheat varieties (C591, C273, 9D, and K78) identified via in silico analysis were analyzed for immunogenicity by measuring T-cell proliferation rate and levels of inflammatory cytokines (Interferon-γ and Tumor Necrosis Factor-α). Peripheral Blood Mononuclear Cells and biopsy derived T-cell lines isolated from four CD patients in complete remission and two controls were stimulated and cultured in the presence of tissue transglutaminase activated pepsin-trypsin (PT) digest of total gliadin extract from test varieties. The immunogenicity was compared with PBW 621, one of the widely cultivated wheat varieties. Phytohaemagglutinin-p was taken as positive control, along with unstimulated cells as negative control. Rate of cell proliferation (0.318, 0.482; 0.369, 0.337), concentration of IFN- γ (107.4, 99.2; 117.9, 99.7 pg/ml), and TNF- α (453.8, 514.2; 463.8, 514.2 pg/ml) was minimum in cultures supplemented with wheat antigen from C273, when compared with other test varieties and unstimulated cells. Significant difference in toxicity levels among different wheat genotypes to stimulate celiac mucosal T-cells and PBMC's was observed; where C273 manifested least immunogenic response amongst the test varieties analyzed.
Collapse
Affiliation(s)
- Jasmine Grover
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, India
| | - Parveen Chhuneja
- School of Agriculture Biotechnology, Punjab Agricultural University, Ludhiana, India
| | - Vandana Midha
- Department of Internal Medicine, Dayanand Medical College, Ludhiana, India
| | - Jean Eric Ghia
- Section of Gastroenterology, Department of Immunology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Dipak Deka
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | | | - Neena Sood
- Department of Pathology, Dayanand Medical College, Ludhiana, India
| | - Ramit Mahajan
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, India
| | - Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, India
| | - Ramneek Verma
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Ekta Bansal
- Department of Biochemistry, Dayanand Medical College, Ludhiana, India
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, India
| |
Collapse
|
58
|
|
59
|
Inflammatory response of gliadin protein isolated from various wheat varieties on human intestinal cell line. J Cereal Sci 2018. [DOI: 10.1016/j.jcs.2018.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
60
|
Medjeber O, Touri K, Rafa H, Djeraba Z, Belkhelfa M, Boutaleb AF, Arroul-Lammali A, Belguendouz H, Touil-Boukoffa C. Ex vivo immunomodulatory effect of ethanolic extract of propolis during Celiac Disease: involvement of nitric oxide pathway. Inflammopharmacology 2018. [PMID: 29516252 DOI: 10.1007/s10787-018-0460-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Celiac Disease (CeD) is a chronic immune-mediated enteropathy, in which dietary gluten induces an inflammatory reaction, predominantly in the duodenum. Propolis is a resinous hive product, collected by honeybees from various plant sources. Propolis is well-known for its anti-inflammatory, anti-oxidant and immunomodulatory effects, due to its major compounds, polyphenols and flavonoids. The aim of our study was to assess the ex vivo effect of ethanolic extract of propolis (EEP) upon the activity and expression of iNOS, along with IFN-γ and IL-10 production in Algerian Celiac patients. In this context, PBMCs isolated from peripheral blood of Celiac patients and healthy controls were cultured with different concentrations of EEP. NO production was measured using the Griess method, whereas quantitation of IFN-γ and IL-10 levels was performed by ELISA. Inducible nitric oxide synthase (iNOS) expression, NFκB and pSTAT-3 activity were analyzed by immunofluorescence assay. Our results showed that PBMCs from Celiac patients produced high levels of NO and IFN-γ compared with healthy controls (HC). Interestingly, EEP reduced significantly, NO and IFN-γ levels and significantly increased IL-10 levels at a concentration of 50 µg/mL. Importantly, EEP downmodulated the iNOS expression as well as the activity of NFκB and pSTAT-3 transcription factors. Altogether, our results highlight the immunomodulatory effect of propolis on NO pathway and on pro-inflammatory cytokines. Therefore, we suggest that propolis may constitute a potential candidate to modulate inflammation during Celiac Disease and has a potential therapeutic value.
Collapse
Affiliation(s)
- Oussama Medjeber
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Kahina Touri
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Hayet Rafa
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Zineb Djeraba
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Mourad Belkhelfa
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | | | - Amina Arroul-Lammali
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Houda Belguendouz
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Chafia Touil-Boukoffa
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria.
| |
Collapse
|
61
|
Sundblad V, Quintar AA, Morosi LG, Niveloni SI, Cabanne A, Smecuol E, Mauriño E, Mariño KV, Bai JC, Maldonado CA, Rabinovich GA. Galectins in Intestinal Inflammation: Galectin-1 Expression Delineates Response to Treatment in Celiac Disease Patients. Front Immunol 2018; 9:379. [PMID: 29545799 PMCID: PMC5837985 DOI: 10.3389/fimmu.2018.00379] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 02/12/2018] [Indexed: 12/21/2022] Open
Abstract
Galectins, a family of animal lectins characterized by their affinity for N-acetyllactosamine-enriched glycoconjugates, modulate several immune cell processes shaping the course of innate and adaptive immune responses. Through interaction with a wide range of glycosylated receptors bearing complex branched N-glycans and core 2-O-glycans, these endogenous lectins trigger distinct signaling programs thereby controling immune cell activation, differentiation, recruitment and survival. Given the unique features of mucosal inflammation and the differential expression of galectins throughout the gastrointestinal tract, we discuss here key findings on the role of galectins in intestinal inflammation, particularly Crohn’s disease, ulcerative colitis, and celiac disease (CeD) patients, as well as in murine models resembling these inflammatory conditions. In addition, we present new data highlighting the regulated expression of galectin-1 (Gal-1), a proto-type member of the galectin family, during intestinal inflammation in untreated and treated CeD patients. Our results unveil a substantial upregulation of Gal-1 accompanying the anti-inflammatory and tolerogenic response associated with gluten-free diet in CeD patients, suggesting a major role of this lectin in favoring resolution of inflammation and restoration of mucosal homeostasis. Thus, a coordinated network of galectins and their glycosylated ligands, exerting either anti-inflammatory or proinflammatory responses, may influence the interplay between intestinal epithelial cells and the highly specialized gut immune system in physiologic and pathologic settings.
Collapse
Affiliation(s)
- Victoria Sundblad
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Amado A Quintar
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Luciano G Morosi
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Sonia I Niveloni
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Ana Cabanne
- Unidad de Patología, Hospital de Gastroenterología, Bonorino Udaondo, Buenos Aires, Argentina
| | - Edgardo Smecuol
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Eduardo Mauriño
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Julio C Bai
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina.,Instituto de Investigaciones, Universidad del Salvador, Buenos Aires, Argentina
| | - Cristina A Maldonado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
62
|
Abstract
Gluten‐related disorders are a complex group of diseases that involve the activation of the immune system triggered by the ingestion of gluten. Among these, celiac disease, with a prevalence of 1 %, is the most investigated, but recently, a new pathology, named nonceliac gluten sensitivity, was reported with a general prevalence of 7 %. Finally, there other less‐prevalent gluten‐related diseases such as wheat allergy, gluten ataxia, and dermatitis herpetiformis (with an overall prevalence of less than 0.1 %). As mentioned, the common molecular trigger is gluten, a complex mixture of storage proteins present in wheat, barley, and a variety of oats that are not fully degraded by humans. The most‐studied protein related to disease is gliadin, present in wheat, which possesses in its sequence many pathological fragments. Despite a lot of effort to treat these disorders, the only effective method is a long‐life gluten‐free diet. This Review summarizes the actual knowledge of gluten‐related disorders from a translational chemistry point of view. We discuss what is currently known from the literature about the interaction of gluten with the gut and the critical host responses it evokes and, finally, connect them to our current and novel molecular understanding of the supramolecular organization of gliadin and the 33‐mer gliadin peptide fragment under physiological conditions.
Collapse
Affiliation(s)
- Karen M Lammers
- Laboratory Immunogenetics, Department of Medical Microbiology and Infection Control VU University Medical Center 1081 Amsterdam Netherlands
| | - Maria G Herrera
- Faculty of Pharmacy and Biochemistry Institute of biological chemistry and Physicochemical CONICET-University of Buenos Aires Junín 956 C1113AAD Buenos Aires Argentina
| | - Veronica I Dodero
- Department of Chemistry, Organic Chemistry III Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| |
Collapse
|
63
|
Vaquero L, Rodríguez-Martín L, León F, Jorquera F, Vivas S. New coeliac disease treatments and their complications. GASTROENTEROLOGIA Y HEPATOLOGIA 2018; 41:191-204. [PMID: 29422237 DOI: 10.1016/j.gastrohep.2017.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/28/2017] [Accepted: 12/14/2017] [Indexed: 02/06/2023]
Abstract
The only accepted treatment for coeliac disease is strict adherence to a gluten-free diet. This type of diet may give rise to reduced patient quality of life with economic and social repercussions. For this reason, dietary transgressions are common and may elicit intestinal damage. Several treatments aimed at different pathogenic targets of coeliac disease have been developed in recent years: modification of gluten to produce non-immunogenic gluten, endoluminal therapies to degrade gluten in the intestinal lumen, increased gluten tolerance, modulation of intestinal permeability and regulation of the adaptive immune response. This review evaluates these coeliac disease treatment lines that are being researched and the treatments that aim to control disease complications like refractory coeliac disease.
Collapse
Affiliation(s)
- Luis Vaquero
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España
| | | | | | - Francisco Jorquera
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España; Instituto de Biomedicina (IBIOMED), Universidad de León, León, España
| | - Santiago Vivas
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España; Instituto de Biomedicina (IBIOMED), Universidad de León, León, España.
| |
Collapse
|
64
|
Coeliac disease in children with type 1 diabetes. THE LANCET CHILD & ADOLESCENT HEALTH 2018; 2:133-143. [DOI: 10.1016/s2352-4642(17)30172-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/13/2017] [Accepted: 10/27/2017] [Indexed: 12/17/2022]
|
65
|
Iacomino G, Aufiero VR, Marena P, Venezia A, Troncone R, Auricchio S, Mazzarella G. Laser Capture Microdissection as a Tool to Study the Mucosal Immune Response in Celiac Disease. Methods Mol Biol 2018; 1723:139-154. [PMID: 29344858 DOI: 10.1007/978-1-4939-7558-7_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Laser capture microdissection (LCM) is a powerful tool for selection and isolation of single cells or compartments from complex primary tissues to perform molecular analyses. Celiac disease is a genetic autoimmune disorder where the ingestion of gluten leads to damage in the small intestine. Increased intraepithelial lymphocytes and the presence of the lamina propria inflammatory infiltrate of the duodenal mucosa is a common part of the disease. These cells promote inflammatory processes through the release of cytokines. Here, we describe the use of LCM and real-time quantitative PCR (RT-qPCR) to analyze cytokine profile information in distinct duodenal mucosa tissue compartments of celiac patients.
Collapse
Affiliation(s)
| | | | | | | | - Riccardo Troncone
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
- European Laboratory for the Investigation of Food-Induced Disease, University of Naples "Federico II", Naples, Italy
| | - Salvatore Auricchio
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
- European Laboratory for the Investigation of Food-Induced Disease, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Mazzarella
- Institute of Food Sciences-CNR, Avellino, Italy.
- European Laboratory for the Investigation of Food-Induced Disease, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
66
|
Zundler S, Neurath MF. Pathogenic T cell subsets in allergic and chronic inflammatory bowel disorders. Immunol Rev 2017; 278:263-276. [DOI: 10.1111/imr.12544] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1; University of Erlangen-Nuremberg; Kussmaul Campus for Medical Research & Translational Research Center; Erlangen Germany
| | - Markus F. Neurath
- Department of Medicine 1; University of Erlangen-Nuremberg; Kussmaul Campus for Medical Research & Translational Research Center; Erlangen Germany
| |
Collapse
|
67
|
Rossi S, Luongo D, Maurano F, Bergamo P, Rossi M. Immunomodulatory activity of recombinant α-gliadin conjugated to cholera toxin in DQ8 transgenic mice. Immunol Lett 2017; 187:47-52. [PMID: 28511837 DOI: 10.1016/j.imlet.2017.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/23/2017] [Accepted: 05/05/2017] [Indexed: 01/29/2023]
Abstract
Coeliac disease (CD) is characterized by an intestinal lesion sustained by an abnormal mucosal T-cell response to wheat gliadin. An immunological approach that is able to suppress this immune response is a perspective worth pursuing. Several strategies of antigen administration have been aimed at the downregulation of pathogenic T-cells. In particular, we previously reported a significant suppression of the systemic cell-mediated response toward wheat gliadin in DQ8 transgenic mice receiving nasally a recombinant α-gliadin. To gain further insight about the cellular mechanisms underlying the tolerogenic properties of this molecule, we analysed different preparations of the recombinant α-gliadin, alone or conjugated to the adjuvant cholera toxin (CT), by in vitro challenge with spleen CD4+ T cells from gliadin-sensitized DQ8 tg mice. We found that a partially purified preparation of recombinant α-gliadin (r-gliadin) induced a significantly higher production of IFN-γ than native gliadin as well as HPLC purified r-gliadin. Interestingly, r-gliadin, but not HPLC purified r-gliadin, stimulated the gliadin-specific expression of IL-10 in CD4+ T cells. No significant cytotoxic effect was induced by r-gliadin in MODE-K cells, a murine model of enterocytes. Notably, a conjugate CT-r-gliadin failed in stimulating IFN-γ, whereas IL-10 secretion was still induced in gliadin-specific CD4+ T cells. In conclusion, our results showed that DCs, pulsed with CT-r-gliadin in vitro, could modulate the ongoing Th1-like T cell response toward wheat gliadin. This finding provides new insight into the design of immunomodulatory protocols potentially useful for CD.
Collapse
Affiliation(s)
| | | | | | | | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
68
|
Moffitt AB, Ondrejka SL, McKinney M, Rempel RE, Goodlad JR, Teh CH, Leppa S, Mannisto S, Kovanen PE, Tse E, Au-Yeung RKH, Kwong YL, Srivastava G, Iqbal J, Yu J, Naresh K, Villa D, Gascoyne RD, Said J, Czader MB, Chadburn A, Richards KL, Rajagopalan D, Davis NS, Smith EC, Palus BC, Tzeng TJ, Healy JA, Lugar PL, Datta J, Love C, Levy S, Dunson DB, Zhuang Y, Hsi ED, Dave SS. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. J Exp Med 2017; 214:1371-1386. [PMID: 28424246 PMCID: PMC5413324 DOI: 10.1084/jem.20160894] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
Enteropathy-associated T cell lymphoma (EATL) is the most common oncologic complication of celiac disease. Moffitt and colleagues identify novel EATL-defining mutations in SETD2, as well as clinically relevant mutations in the JAK-STAT pathway. Enteropathy-associated T cell lymphoma (EATL) is a lethal, and the most common, neoplastic complication of celiac disease. Here, we defined the genetic landscape of EATL through whole-exome sequencing of 69 EATL tumors. SETD2 was the most frequently silenced gene in EATL (32% of cases). The JAK-STAT pathway was the most frequently mutated pathway, with frequent mutations in STAT5B as well as JAK1, JAK3, STAT3, and SOCS1. We also identified mutations in KRAS, TP53, and TERT. Type I EATL and type II EATL (monomorphic epitheliotropic intestinal T cell lymphoma) had highly overlapping genetic alterations indicating shared mechanisms underlying their pathogenesis. We modeled the effects of SETD2 loss in vivo by developing a T cell–specific knockout mouse. These mice manifested an expansion of γδ T cells, indicating novel roles for SETD2 in T cell development and lymphomagenesis. Our data render the most comprehensive genetic portrait yet of this uncommon but lethal disease and may inform future classification schemes.
Collapse
Affiliation(s)
- Andrea B Moffitt
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Sarah L Ondrejka
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Matthew McKinney
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Rachel E Rempel
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - John R Goodlad
- Haematological Malignancy Diagnostic Service, St. James's University Hospital, Leeds LS9 7TF, England, UK
| | - Chun Huat Teh
- Haematology Department, Western General Hospital, Edinburgh EH14 1TY, Scotland, UK
| | - Sirpa Leppa
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Susanna Mannisto
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Panu E Kovanen
- HUSLAB and Medicum, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Eric Tse
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Yok-Lam Kwong
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Javeed Iqbal
- University of Nebraska Medical Center, Omaha, NE 68198
| | - Jiayu Yu
- University of Nebraska Medical Center, Omaha, NE 68198
| | | | - Diego Villa
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Randy D Gascoyne
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Jonathan Said
- University of California, Los Angeles, Los Angeles, CA 90095
| | | | - Amy Chadburn
- Presbyterian Hospital, Pathology and Cell Biology, Cornell University, New York, NY 10065
| | | | | | - Nicholas S Davis
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Eileen C Smith
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Brooke C Palus
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Tiffany J Tzeng
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Jane A Healy
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Patricia L Lugar
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - Jyotishka Datta
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Cassandra Love
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Shawn Levy
- Hudson Alpha Institute for Biotechnology, Huntsville, AL 35806
| | - David B Dunson
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Yuan Zhuang
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710
| | - Eric D Hsi
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sandeep S Dave
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| |
Collapse
|
69
|
Bouziat R, Hinterleitner R, Brown JJ, Stencel-Baerenwald JE, Ikizler M, Mayassi T, Meisel M, Kim SM, Discepolo V, Pruijssers AJ, Ernest JD, Iskarpatyoti JA, Costes LMM, Lawrence I, Palanski BA, Varma M, Zurenski MA, Khomandiak S, McAllister N, Aravamudhan P, Boehme KW, Hu F, Samsom JN, Reinecker HC, Kupfer SS, Guandalini S, Semrad CE, Abadie V, Khosla C, Barreiro LB, Xavier RJ, Ng A, Dermody TS, Jabri B. Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease. Science 2017; 356:44-50. [PMID: 28386004 PMCID: PMC5506690 DOI: 10.1126/science.aah5298] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 02/22/2017] [Indexed: 12/11/2022]
Abstract
Viral infections have been proposed to elicit pathological processes leading to the initiation of T helper 1 (TH1) immunity against dietary gluten and celiac disease (CeD). To test this hypothesis and gain insights into mechanisms underlying virus-induced loss of tolerance to dietary antigens, we developed a viral infection model that makes use of two reovirus strains that infect the intestine but differ in their immunopathological outcomes. Reovirus is an avirulent pathogen that elicits protective immunity, but we discovered that it can nonetheless disrupt intestinal immune homeostasis at inductive and effector sites of oral tolerance by suppressing peripheral regulatory T cell (pTreg) conversion and promoting TH1 immunity to dietary antigen. Initiation of TH1 immunity to dietary antigen was dependent on interferon regulatory factor 1 and dissociated from suppression of pTreg conversion, which was mediated by type-1 interferon. Last, our study in humans supports a role for infection with reovirus, a seemingly innocuous virus, in triggering the development of CeD.
Collapse
Affiliation(s)
- Romain Bouziat
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Reinhard Hinterleitner
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Judy J Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer E Stencel-Baerenwald
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mine Ikizler
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Toufic Mayassi
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Marlies Meisel
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Sangman M Kim
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Valentina Discepolo
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Department of Translational Medical Sciences, Section of Pediatrics, University of Naples Federico II, and CeInGe-Biotecnologie Avanzate, Naples, Italy
| | - Andrea J Pruijssers
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Jason A Iskarpatyoti
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Léa M M Costes
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Ian Lawrence
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Brad A Palanski
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Mukund Varma
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Matthew A Zurenski
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Solomiia Khomandiak
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicole McAllister
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pavithra Aravamudhan
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Karl W Boehme
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fengling Hu
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Hans-Christian Reinecker
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sonia S Kupfer
- Department of Medicine, University of Chicago, Chicago, IL, USA
- University of Chicago Celiac Disease Center, University of Chicago, Chicago, IL, USA
| | - Stefano Guandalini
- University of Chicago Celiac Disease Center, University of Chicago, Chicago, IL, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Carol E Semrad
- Department of Medicine, University of Chicago, Chicago, IL, USA
- University of Chicago Celiac Disease Center, University of Chicago, Chicago, IL, USA
| | - Valérie Abadie
- Department of Microbiology, Infectiology, and Immunology, University of Montreal, and the Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Stanford ChEM-H, Stanford University, Stanford, California, USA
| | - Luis B Barreiro
- Department of Genetics, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Ramnik J Xavier
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aylwin Ng
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Terence S Dermody
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- University of Chicago Celiac Disease Center, University of Chicago, Chicago, IL, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Chicago, Chicago, IL, USA
- Department of Pathology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
70
|
Escudero-Hernández C, Peña AS, Bernardo D. Immunogenetic Pathogenesis of Celiac Disease and Non-celiac Gluten Sensitivity. Curr Gastroenterol Rep 2017; 18:36. [PMID: 27216895 DOI: 10.1007/s11894-016-0512-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Celiac disease is the most common oral intolerance in Western countries. It results from an immune response towards gluten proteins from certain cereals in genetically predisposed individuals (HLA-DQ2 and/or HLA-DQ8). Its pathogenesis involves the adaptive (HLA molecules, transglutaminase 2, dendritic cells, and CD4(+) T-cells) and the innate immunity with an IL-15-mediated response elicited in the intraepithelial compartment. At present, the only treatment is a permanent strict gluten-free diet (GFD). Multidisciplinary studies have provided a deeper insight of the genetic and immunological factors and their interaction with the microbiota in the pathogenesis of the disease. Similarly, a better understanding of the composition of the toxic gluten peptides has improved the ways to detect them in food and drinks and how to monitor GFD compliance via non-invasive approaches. This review, therefore, addresses the major findings obtained in the last few years including the re-discovery of non-celiac gluten sensitivity.
Collapse
Affiliation(s)
- Celia Escudero-Hernández
- Mucosal Immunology Laboratory, IBGM, Facultad de Medicina, Dpto. Pediatría e Inmunología, University of Valladolid-Consejo Superior de Investigaciones Científicas, (4th floor) Av. Ramón y Cajal 7, 47005, Valladolid, Spain
| | - Amado Salvador Peña
- VU Medical Center Amsterdam, Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108 Room 10E65, 1081 HZ, Amsterdam, The Netherlands
| | - David Bernardo
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, 28006, Spain.
| |
Collapse
|
71
|
CD4 T-cell cytokines synergize to induce proliferation of malignant and nonmalignant innate intraepithelial lymphocytes. Proc Natl Acad Sci U S A 2017; 114:E980-E989. [PMID: 28049849 DOI: 10.1073/pnas.1620036114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Refractory celiac disease type II (RCDII) is a severe complication of celiac disease (CD) characterized by the presence of an enlarged clonal population of innate intraepithelial lymphocytes (IELs) lacking classical B-, T-, and natural killer (NK)-cell lineage markers (Lin-IELs) in the duodenum. In ∼50% of patients with RCDII, these Lin-IELs develop into a lymphoma for which no effective treatment is available. Current evidence indicates that the survival and expansion of these malignant Lin-IELs is driven by epithelial cell-derived IL-15. Like CD, RCDII is strongly associated with HLA-DQ2, suggesting the involvement of HLA-DQ2-restricted gluten-specific CD4+ T cells. We now show that gluten-specific CD4+ T cells isolated from CD duodenal biopsy specimens produce cytokines able to trigger proliferation of malignant Lin-IEL lines as powerfully as IL-15. Furthermore, we identify TNF, IL-2, and IL-21 as CD4+ T-cell cytokines that synergistically mediate this effect. Like IL-15, these cytokines were found to increase the phosphorylation of STAT5 and Akt and transcription of antiapoptotic mediator bcl-xL Several small-molecule inhibitors targeting the JAK/STAT pathway blocked proliferation elicited by IL-2 and IL-15, but only an inhibitor targeting the PI3K/Akt/mTOR pathway blocked proliferation induced by IL-15 as well as the CD4+ T-cell cytokines. Confirming and extending these findings, TNF, IL-2, and IL-21 also synergistically triggered the proliferation of freshly isolated Lin-IELs and CD3-CD56+ IELs (NK-IELs) from RCDII as well as non-RCDII duodenal biopsy specimens. These data provide evidence implicating CD4+ T-cell cytokines in the pathogenesis of RCDII. More broadly, they suggest that adaptive immune responses can contribute to innate IEL activation during mucosal inflammation.
Collapse
|
72
|
Abstract
Celiac disease is an autoimmune disorder induced by gluten in genetically susceptible individuals. It can result in intraintestinal and extraintestinal manifestations of disease including diarrhea, weight loss, anemia, osteoporosis, or lymphoma. Diagnosis of celiac disease is made through initial serologic testing and then confirmed by histopathologic examination of duodenal biopsies. Generally celiac disease is a benign disorder with a good prognosis in those who adhere to a gluten-free diet. However, in refractory disease, complications may develop that warrant additional testing with more advanced radiologic and endoscopic methods. This article reviews the current strategy to diagnose celiac disease and the newer modalities to assess for associated complications.
Collapse
Affiliation(s)
- Sarah Shannahan
- Division of Internal Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Daniel A Leffler
- Division of Gastroenterology, The Celiac Center, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
73
|
Plugis NM, Palanski BA, Weng CH, Albertelli M, Khosla C. Thioredoxin-1 Selectively Activates Transglutaminase 2 in the Extracellular Matrix of the Small Intestine: IMPLICATIONS FOR CELIAC DISEASE. J Biol Chem 2016; 292:2000-2008. [PMID: 28003361 DOI: 10.1074/jbc.m116.767988] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/13/2016] [Indexed: 11/06/2022] Open
Abstract
Transglutaminase 2 (TG2) catalyzes transamidation or deamidation of its substrates and is ordinarily maintained in a catalytically inactive state in the intestine and other organs. Aberrant TG2 activity is thought to play a role in celiac disease, suggesting that a better understanding of TG2 regulation could help to elucidate the mechanistic basis of this malady. Structural and biochemical analysis has led to the hypothesis that extracellular TG2 activation involves reduction of an allosteric disulfide bond by thioredoxin-1 (TRX), but cellular and in vivo evidence for this proposal is lacking. To test the physiological relevance of this hypothesis, we first showed that macrophages exposed to pro-inflammatory stimuli released TRX in sufficient quantities to activate their extracellular pools of TG2. By using the C35S mutant of TRX, which formed a metastable mixed disulfide bond with TG2, we demonstrated that these proteins specifically recognized each other in the extracellular matrix of fibroblasts. When injected into mice and visualized with antibodies, we observed the C35S TRX mutant bound to endogenous TG2 as its principal protein partner in the small intestine. Control experiments showed no labeling of TG2 knock-out mice. Intravenous administration of recombinant TRX in wild-type mice, but not TG2 knock-out mice, led to a rapid rise in intestinal transglutaminase activity in a manner that could be inhibited by small molecules targeting TG2 or TRX. Our findings support the potential pathophysiological relevance of TRX in celiac disease and establish the Cys370-Cys371 disulfide bond of TG2 as one of clearest examples of an allosteric disulfide bond in mammals.
Collapse
Affiliation(s)
- Nicholas M Plugis
- From the Department of Chemistry, Stanford University, Stanford, California 94305
| | - Brad A Palanski
- From the Department of Chemistry, Stanford University, Stanford, California 94305
| | - Chih-Hisang Weng
- From the Department of Chemistry, Stanford University, Stanford, California 94305; the School of Medicine, Stanford University, Stanford, California 94305; the Medical Science Training Program, Stanford University, Stanford, California 94305
| | - Megan Albertelli
- Department of Comparative Medicine, Stanford University, Stanford, California 94305
| | - Chaitan Khosla
- From the Department of Chemistry, Stanford University, Stanford, California 94305; Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford ChEM-H, Stanford University, Stanford, California 94305.
| |
Collapse
|
74
|
Properties of Gluten Intolerance: Gluten Structure, Evolution, Pathogenicity and Detoxification Capabilities. Nutrients 2016; 8:nu8100644. [PMID: 27763541 PMCID: PMC5084031 DOI: 10.3390/nu8100644] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 09/30/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022] Open
Abstract
Theterm gluten intolerance may refer to three types of human disorders: autoimmune celiac disease (CD), allergy to wheat and non-celiac gluten sensitivity (NCGS). Gluten is a mixture of prolamin proteins present mostly in wheat, but also in barley, rye and oat. Gluten can be subdivided into three major groups: S-rich, S-poor and high molecular weight proteins. Prolamins within the groups possess similar structures and properties. All gluten proteins are evolutionarily connected and share the same ancestral origin. Gluten proteins are highly resistant to hydrolysis mediated by proteases of the human gastrointestinal tract. It results in emergence of pathogenic peptides, which cause CD and allergy in genetically predisposed people. There is a hierarchy of peptide toxicity and peptide recognition by T cells. Nowadays, there are several ways to detoxify gluten peptides: the most common is gluten-free diet (GFD), which has proved its effectiveness; prevention programs, enzymatic therapy, correction of gluten pathogenicity pathways and genetically modified grains with reduced immunotoxicity. A deep understanding of gluten intolerance underlying mechanisms and detailed knowledge of gluten properties may lead to the emergence of novel effective approaches for treatment of gluten-related disorders.
Collapse
|
75
|
Transglutaminase 2-specific coeliac disease autoantibodies induce morphological changes and signs of inflammation in the small-bowel mucosa of mice. Amino Acids 2016; 49:529-540. [PMID: 27503559 DOI: 10.1007/s00726-016-2306-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/30/2016] [Indexed: 01/28/2023]
Abstract
Coeliac disease is hallmarked by an abnormal immune reaction against ingested wheat-, rye- and barley-derived gluten and the presence of transglutaminase 2 (TG2)-targeted autoantibodies. The small-bowel mucosal damage characteristic of the disorder develops gradually from normal villus morphology to inflammation and finally to villus atrophy with crypt hyperplasia. Patients with early-stage coeliac disease have TG2-autoantibodies present in serum and small-intestinal mucosa and they may already suffer from abdominal symptoms before the development of villus atrophy. Previously, we have shown that intraperitoneal injections of coeliac patient-derived sera or purified immunoglobulin fraction into mice induce a condition mimicking early-stage coeliac disease. In the current study, we sought to establish whether recombinantly produced patient-derived TG2-targeted autoantibodies are by themselves sufficient for the development of such an experimentally induced condition in immune-compromised mice. Interestingly, mice injected with coeliac patient TG2-antibodies had altered small-intestinal mucosal morphology, increased lamina propria cellular infiltration and disease-specific autoantibodies deposited in the small bowel, but did not evince clinical features of the disease. Thus, coeliac patient-derived TG2-specific autoantibodies seem to be sufficient for the induction of subtle small-bowel mucosal alterations in mice, but the development of clinical features probably requires additional factors such as other antibody populations relevant in coeliac disease.
Collapse
|
76
|
Mast cell activation disease and the modern epidemic of chronic inflammatory disease. Transl Res 2016; 174:33-59. [PMID: 26850903 DOI: 10.1016/j.trsl.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
Abstract
A large and growing portion of the human population, especially in developed countries, suffers 1 or more chronic, often quite burdensome ailments which either are overtly inflammatory in nature or are suspected to be of inflammatory origin, but for which investigations to date have failed to identify specific causes, let alone unifying mechanisms underlying the multiple such ailments that often afflict such patients. Relatively recently described as a non-neoplastic cousin of the rare hematologic disease mastocytosis, mast cell (MC) activation syndrome-suspected to be of greatly heterogeneous, complex acquired clonality in many cases-is a potential underlying/unifying explanation for a diverse assortment of inflammatory ailments. A brief review of MC biology and how aberrant primary MC activation might lead to such a vast range of illness is presented.
Collapse
|
77
|
Cappello M, Morreale GC, Licata A. Elderly Onset Celiac Disease: A Narrative Review. CLINICAL MEDICINE INSIGHTS. GASTROENTEROLOGY 2016; 9:41-9. [PMID: 27486350 PMCID: PMC4965017 DOI: 10.4137/cgast.s38454] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 12/16/2022]
Abstract
Celiac sprue is a chronic disease, which usually occurs in children and young adults. However, it can develop in any age group, and the prevalence is increasing even in the elderly population. The atypical patterns of clinical presentation in this age group sometimes can cause a delay in diagnosis. Given the lower sensitivity and specificity of serological tests in the aged population, clinical suspect often arises in the presence of complications (autoimmune disorders, fractures, and finally, malignancy) and must be supported by endoscopic and imaging tools. In this review, we highlight the incidence and prevalence of celiac disease in the elderly, the patterns of clinical presentation, diagnosis, and the most frequent complications, with the aim of increasing awareness and reducing the diagnostic delay of celiac disease even in the elderly population.
Collapse
Affiliation(s)
- Maria Cappello
- Gastroenterology and Hepatology Section, DIBIMIS, University of Palermo School of Medicine, Palermo, Italy
| | - Gaetano C Morreale
- Gastroenterology and Hepatology Section, DIBIMIS, University of Palermo School of Medicine, Palermo, Italy
| | - Anna Licata
- Gastroenterology and Hepatology Section, DIBIMIS, University of Palermo School of Medicine, Palermo, Italy
| |
Collapse
|
78
|
Yang G, Zhang B, Huang W, Zhang N, Dong F, Jing L, Wang M, Liu Y, Guo C, Pan H, Wei X, Jing C. Systematic review and meta-analysis of the association between IL18RAP rs917997 and CCR3 rs6441961 polymorphisms with celiac disease risks. Expert Rev Gastroenterol Hepatol 2016; 9:1327-38. [PMID: 26289103 DOI: 10.1586/17474124.2015.1075880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES We performed a systematic review and meta-analysis to estimate the polymorphism effects of IL18RAP and CCR3 on celiac disease susceptibility. METHODS PubMed and Web of Science databases were searched (to June 2015) on IL18RAP rs917997 and CCR3 rs6441961 polymorphisms. RESULTS The meta-analysis included 16 and 7 studies for rs917997 and rs6441961, respectively. The minor risk A allele at both rs917997 and rs6441961 carried risks (odds ratios) of 1.24 (95% CI 1.18-1.31) and 1.21 (95% CI 1.12-1.31), respectively. These alleles contributed to increase risks in all celiac disease patients by 5.04 and 6.35%. The estimated lambdas were 0.73 and 0.51, suggesting that an additive model would be the best choice for both gene effects. CONCLUSIONS This meta-analysis provides robust estimates that IL18RAP rs917997 and CCR3 rs6441961 are potential risk factors for celiac disease in European populations. Studies are needed to confirm these findings in different ethnicities.
Collapse
Affiliation(s)
- Guang Yang
- a 1 Department of Parasitology, School of Medicine, Jinan University, Guangzhou, China
| | - Baohuan Zhang
- a 1 Department of Parasitology, School of Medicine, Jinan University, Guangzhou, China
| | - Weihuang Huang
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Na Zhang
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Fang Dong
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Lipeng Jing
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Man Wang
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yang Liu
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Congcong Guo
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hongwei Pan
- c 3 Department of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiangcai Wei
- d 4 Family Planning Research Institute of Guangdong, Guangzhou, China
| | - Chunxia Jing
- b 2 Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
79
|
Ayala-Fontánez N, Soler DC, McCormick TS. Current knowledge on psoriasis and autoimmune diseases. PSORIASIS-TARGETS AND THERAPY 2016; 6:7-32. [PMID: 29387591 PMCID: PMC5683130 DOI: 10.2147/ptt.s64950] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Psoriasis is a prevalent, chronic inflammatory disease of the skin, mediated by crosstalk between epidermal keratinocytes, dermal vascular cells, and immunocytes such as antigen presenting cells (APCs) and T cells. Exclusive cellular “responsibility” for the induction and maintenance of psoriatic plaques has not been clearly defined. Increased proliferation of keratinocytes and endothelial cells in conjunction with APC/T cell/monocyte/macrophage inflammation leads to the distinct epidermal and vascular hyperplasia that is characteristic of lesional psoriatic skin. Despite the identification of numerous susceptibility loci, no single genetic determinant has been identified as responsible for the induction of psoriasis. Thus, numerous other triggers of disease, such as environmental, microbial and complex cellular interactions must also be considered as participants in the development of this multifactorial disease. Recent advances in therapeutics, especially systemic so-called “biologics” have provided new hope for identifying the critical cellular targets that drive psoriasis pathogenesis. Recent recognition of the numerous co-morbidities and other autoimmune disorders associated with psoriasis, including inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus suggest common signaling elements and cellular mediators may direct disease pathogenesis. In this review, we discuss common cellular pathways and participants that mediate psoriasis and other autoimmune disorders that share these cellular signaling pathways.
Collapse
Affiliation(s)
- Nilmarie Ayala-Fontánez
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.,The Murdough Family Center for Psoriasis, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - David C Soler
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.,The Murdough Family Center for Psoriasis, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.,The Murdough Family Center for Psoriasis, University Hospitals Case Medical Center, Cleveland, OH, USA
| |
Collapse
|
80
|
Khaleghi S, Ju JM, Lamba A, Murray JA. The potential utility of tight junction regulation in celiac disease: focus on larazotide acetate. Therap Adv Gastroenterol 2016; 9:37-49. [PMID: 26770266 PMCID: PMC4699279 DOI: 10.1177/1756283x15616576] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Celiac disease (CD) is a common chronic immune disease triggered by gluten. Gliadin peptides pass through the epithelial layers, either paracellularly or transcellularly, to launch a potent adaptive immune response in the lamina propria. This aberrant immune response leads to diverse gastrointestinal and extra-gastrointestinal symptoms. Currently, the only treatment for CD is a strict lifelong adherence to a gluten-free diet (GFD), which can be challenging. An early effect of gluten in CD is an increase in gut permeability. Larazotide acetate, also known as AT-1001, is a synthetic peptide developed as a permeability regulator primarily targeting CD. In vitro studies indicate that larazotide acetate is capable of inhibiting the actin rearrangement caused by gliadin and clinical studies have been conducted using this peptide as a therapy for CD.
Collapse
Affiliation(s)
- Shahryar Khaleghi
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Josephine M. Ju
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Abhinav Lamba
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Joseph A. Murray
- Professor of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| |
Collapse
|
81
|
Iacomino G, Marano A, Stillitano I, Aufiero VR, Iaquinto G, Schettino M, Masucci A, Troncone R, Auricchio S, Mazzarella G. Celiac disease: role of intestinal compartments in the mucosal immune response. Mol Cell Biochem 2016; 411:341-349. [PMID: 26541753 DOI: 10.1007/s11010-015-2596-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/29/2015] [Indexed: 02/06/2023]
Abstract
Different approaches have been used to study the pattern of cytokines in celiac disease (CD). Laser capture microdissection (LCM) is a powerful tool for the isolation of specific tissue compartments. We aimed to investigate the mucosal immune response that takes place in different intestinal compartments of CD patients, dissected by LCM, analyzing cytokine expression profile. Frozen section of jejunum was obtained from 15 untreated CD and 15 control. Surface epithelium and lamina propria compartment were isolated by LCM. RNA from each LCM sample was extracted and, after a retrotranscription step, messenger RNA levels for MxA, IL-15, TNF-α, IFN-γ, IL-17α, IL-21, IL-10, and TGF-β were determined by quantitative reverse transcriptase-PCR. Increased gene expression levels of MxA, IL-15, TNF-α, IL-10, and TGF-β was observed in the surface epithelium of untreated CD with respect to control. Furthermore, all the cytokines investigated were upregulated in the lamina propria of untreated CD as compared to control. Within the untreated CD group the expression of IL-15 was higher, in the surface epithelium than in the lamina propria, whereas the expression levels of IL-17 and IL-21 were higher in the lamina propria than in the surface epithelium. Finally, high levels of IL-10 and TGF-β were detected in both compartments of untreated CD biopsies. In CD, surface epithelium and lamina propria compartments, play a prominent role in determining innate and adaptive immunity, respectively. Conversely, surface epithelium and lamina propria produce high levels of anti-inflammatory cytokines, suggesting that both compartments are involved in the immunoregulatory response.
Collapse
Affiliation(s)
- Giuseppe Iacomino
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy
| | - Angela Marano
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy
| | - Ilaria Stillitano
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy
| | | | - Gaetano Iaquinto
- Gastroenterology and Digestive Endoscopy Service, S. Rita Clinic, via Appia, 83042, Atripalda, Italy
| | - Michele Schettino
- Gastroenterology and Digestive Endoscopy Service, San G. Moscati Hospital, Contrada Amoretta, 83100, Avellino, Italy
| | - Armando Masucci
- Department of Preventive Medicine San G. Moscati Hospital, Contrada Amoretta, 83100, Avellino, Italy
| | - Riccardo Troncone
- Department of Translational Medicine-Pediatric Section, University of Naples "Federico II", Naples, Italy
- European Laboratory for the Investigation of Food-Induced Disease (ELFID), University of Naples "Federico II", via Pansini, 80131, Naples, Italy
| | - Salvatore Auricchio
- Department of Translational Medicine-Pediatric Section, University of Naples "Federico II", Naples, Italy
- European Laboratory for the Investigation of Food-Induced Disease (ELFID), University of Naples "Federico II", via Pansini, 80131, Naples, Italy
| | - Giuseppe Mazzarella
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy.
- European Laboratory for the Investigation of Food-Induced Disease (ELFID), University of Naples "Federico II", via Pansini, 80131, Naples, Italy.
| |
Collapse
|
82
|
Borrelli M, Gianfrani C, Lania G, Aitoro R, Ferrara K, Nanayakkara M, Ponticelli D, Zanzi D, Discepolo V, Vitale S, Barone MV, Troncone R, Auricchio R, Maglio M. In the Intestinal Mucosa of Children With Potential Celiac Disease IL-21 and IL-17A are Less Expressed than in the Active Disease. Am J Gastroenterol 2016; 111:134-44. [PMID: 26753888 DOI: 10.1038/ajg.2015.390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 11/11/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Potential celiac disease (CD) patients are at an increased risk to developing CD as indicated by positive CD-associated serology. We investigated in duodenal mucosa of such patients the presence of both IL-21 and IL-17A and the role of gliadin peptides and IL-15 in their expression. METHODS Duodenal biopsies from 76 active CD, 90 potential CD, and 58 control patients were analyzed for IL-21 and/or IL-17A production by quantitative real-time PCR, immunohistochemistry, flow cytometry, and ELISA. The presence of IL-21 receptor was investigated by western blot. Potential CD duodenal fragments were cultured with gliadin peptides (PTG) and/or IL-15 and the expression/production of IL-21 and IL-17A assessed by quantitative real-time PCR and by immunohistochemistry. RESULTS In potential CD, IL-21 was lower than in active CD, in terms of RNA expression (P<0.01), density of lamina propria (LP) IL-21(+) cells (P<0.05), and protein secretion (P<0.05). Also, IL-21R was weakly detectable in potential CD. Several LP cell types produced IL-21 in CD. In potential CD, CD4(+)IL-21(+) cells increased after PMA-ionomycin stimulation and co-produced IFN-γ but not IL-17A. After 24 hours of culture stimulation with PTG, IL-21-producing cells increased but not the ones producing IL-17A. This increase was further enhanced by the addition of IL-15 to culture medium. CONCLUSIONS In potential CD, IL-21 is less expressed than in active CD; however, IL-21-producing cells are present and prone to respond after specific stimuli. This suggests a key role of IL-21 in the progression of mucosal damage in CD.
Collapse
Affiliation(s)
- Melissa Borrelli
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | | | - Giuliana Lania
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Rosita Aitoro
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Katia Ferrara
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Merlin Nanayakkara
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Domenico Ponticelli
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Delia Zanzi
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Valentina Discepolo
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Serena Vitale
- Institute of Protein Biochemistry, CNR, Naples, Italy
| | - Maria Vittoria Barone
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Riccardo Troncone
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Renata Auricchio
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Mariantonia Maglio
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| |
Collapse
|
83
|
Sams A, Hawks J. Celiac disease as a model for the evolution of multifactorial disease in humans. Hum Biol 2015; 86:19-36. [PMID: 25401984 DOI: 10.3378/027.086.0102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2013] [Indexed: 11/05/2022]
Abstract
Celiac disease (CD) is a multifactorial chronic inflammatory condition that results in injury of the mucosal lining of the small intestine upon ingestion of wheat gluten and related proteins from barley and rye. Although the exact mechanisms leading to CD are not fully understood, the genetic basis of CD has been relatively well characterized. In this review we briefly review the history of discovery, clinical presentation, pathophysiology, and current understanding of the genetics underlying CD risk. Then, we discuss what is known about the current distribution and evolutionary history of genes underlying CD risk in light of other evolutionary models of disease. Specifically, we conclude that the set of loci underlying CD risk did not cohesively evolve as a response to a single past selection event such as the development of agriculture. Rather, deterministic and stochastic evolutionary processes have both contributed to the present distribution of variation in CD risk loci. Selection has shaped some components of this network, but this selection appears to have occurred at different points in the past. Other parts of the CD risk network have likely arisen due to stochastic processes such as genetic drift.
Collapse
Affiliation(s)
- Aaron Sams
- Cornell University, Ithaca, New York, USA
| | - John Hawks
- University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
84
|
Quinn EM, Coleman C, Molloy B, Dominguez Castro P, Cormican P, Trimble V, Mahmud N, McManus R. Transcriptome Analysis of CD4+ T Cells in Coeliac Disease Reveals Imprint of BACH2 and IFNγ Regulation. PLoS One 2015; 10:e0140049. [PMID: 26444573 PMCID: PMC4596691 DOI: 10.1371/journal.pone.0140049] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/21/2015] [Indexed: 12/16/2022] Open
Abstract
Genetic studies have to date identified 43 genome wide significant coeliac disease susceptibility (CD) loci comprising over 70 candidate genes. However, how altered regulation of such disease associated genes contributes to CD pathogenesis remains to be elucidated. Recently there has been considerable emphasis on characterising cell type specific and stimulus dependent genetic variants. Therefore in this study we used RNA sequencing to profile over 70 transcriptomes of CD4+ T cells, a cell type crucial for CD pathogenesis, in both stimulated and resting samples from individuals with CD and unaffected controls. We identified extensive transcriptional changes across all conditions, with the previously established CD gene IFNy the most strongly up-regulated gene (log2 fold change 4.6; Padjusted = 2.40x10-11) in CD4+ T cells from CD patients compared to controls. We show a significant correlation of differentially expressed genes with genetic studies of the disease to date (Padjusted = 0.002), and 21 CD candidate susceptibility genes are differentially expressed under one or more of the conditions used in this study. Pathway analysis revealed significant enrichment of immune related processes. Co-expression network analysis identified several modules of coordinately expressed CD genes. Two modules were particularly highly enriched for differentially expressed genes (P<2.2x10-16) and highlighted IFNy and the genetically associated transcription factor BACH2 which showed significantly reduced expression in coeliac samples (log2FC -1.75; Padjusted = 3.6x10-3) as key regulatory genes in CD. Genes regulated by BACH2 were very significantly over-represented among our differentially expressed genes (P<2.2x10-16) indicating that reduced expression of this master regulator of T cell differentiation promotes a pro-inflammatory response and strongly corroborates genetic evidence that BACH2 plays an important role in CD pathogenesis.
Collapse
Affiliation(s)
- Emma M. Quinn
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Ciara Coleman
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Ben Molloy
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Patricia Dominguez Castro
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Paul Cormican
- Animal and Bioscience Research Department, Grange Research Centre, Teagasc, Dunsany, Ireland
| | - Valerie Trimble
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Nasir Mahmud
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
| | - Ross McManus
- Department of Clinical Medicine, Trinity College Dublin, Trinity Centre, St James’s Hospital, Dublin, 8, Ireland
- * E-mail:
| |
Collapse
|
85
|
Abstract
Celiac disease is a multisystem immune based disorder that is triggered by the ingestion of gluten in genetically susceptible individuals. The prevalence of celiac disease has risen in recent decades and is currently about 1% in most Western populations. The reason for this rise is unknown, although environmental factors related to the hygiene hypothesis are suspected. The pathophysiology of celiac disease involves both the innate and adaptive immune response to dietary gluten. Clinical features are diverse and include gastrointestinal symptoms, metabolic bone disease, infertility, and many other manifestations. Although a gluten-free diet is effective in most patients, this diet can be burdensome and can limit quality of life; consequently, non-dietary therapies are at various stages of development. This review also covers non-celiac gluten sensitivity. The pathophysiology of this clinical phenotype is poorly understood, but it is a cause of increasing interest in gluten-free diets in the general population.
Collapse
Affiliation(s)
- Benjamin Lebwohl
- Celiac Disease Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Medical Epidemiology and Biostatistics, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden Department of Pediatrics, Örebro University Hospital, Sweden
| | - Peter H R Green
- Celiac Disease Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| |
Collapse
|
86
|
Ortiz Sánchez JP, Mata Haro V, Cabrera Chávez F, Calderón de la Barca AM. Prolamins of maize and wheat differentially affect intestinal cells both in biopsies of celiac patients and CACO-2 cell line. FOOD AGR IMMUNOL 2015. [DOI: 10.1080/09540105.2015.1086316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
87
|
Onset of Thrombin Generation Occurs More Rapidly in Pediatric Patients With Celiac Disease. J Pediatr Gastroenterol Nutr 2015; 61:230-3. [PMID: 25793906 DOI: 10.1097/mpg.0000000000000786] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Celiac disease (CD) is a risk factor for venous thromboembolism (VTE) and stroke, but the mechanisms are unclear. Continuous measurement of thrombin generation in plasma is a feasible way to detect hypercoagulable changes. The aim of this pilot study was to investigate thrombin generation in pediatric patients with CD compared with pediatric controls. METHODS Plasma samples were collected from 19 pediatric patients with CD and 20 healthy controls. In each patient diagnosed as having CD, thrombin generation was determined twice by means of calibrated automated thrombography. The first measurement was undertaken when CD was diagnosed; the second measurement was undertaken after normalization of their IgA antitissue transglutaminase antibody (tTG-Ab) titers following a gluten-free diet. In the controls, measurement for TTG-Ab and thrombin generation was undertaken once during recruitment. RESULTS Patients with CD at diagnosis showed a significantly shorter lag time compared with controls (P < 0.001) and a shorter time-to-peak compared with controls (P < 0.02). These differences were no longer detectable after normalization of TTG-Ab values. The overall amount of generated thrombin, represented by the endogenous thrombin potential (ETP), showed no significant difference between the study groups. CONCLUSIONS Our results show that alterations in coagulation can be found in untreated CD that may help to explain the described increased risk of stroke or VTE. A shorter lag time in patients with untreated CD indicates a more rapid onset of thrombin generation as a sign of hypercoagulability. ETP, the best predictive parameter for thromboembolic disease, however, was not altered.
Collapse
|
88
|
Gujral N, Suh JW, Sunwoo HH. Effect of anti-gliadin IgY antibody on epithelial intestinal integrity and inflammatory response induced by gliadin. BMC Immunol 2015; 16:41. [PMID: 26156219 PMCID: PMC4495697 DOI: 10.1186/s12865-015-0104-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 06/24/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pepsin-trypsin resistant gliadin (PT-gliadin) promotes intestinal tissue inflammation and increases paracellular permeability of immunogenic gliadin peptides into the lamina propria. This leads to the complications seen in the pathogenesis of celiac disease (CD). In this study, specific anti-gliadin IgY antibody was produced and evaluated for its efficacy on gliadin induced intestinal integrity impairment and proinflammatory effects on intestinal epithelial (Caco-2) cell culture model for CD. METHODS Caco-2 (passages 20-24) monolayers were subjected to 7 experimental conditions (n=3 each): phosphate buffered saline (PBS; control), pancreatic digested-casein (PD-casein; negative control), PT-gliadin (positive control), non-specific IgY with PT-gliadin, and anti-wheat gliadin IgY with PT-gliadin at a ratio of 1:6,000, 1:3,000 and 1:1,500. Caco-2 monolayers were then evaluated for effects of gliadin and/or anti-wheat gliadin IgY after 24 h exposure. Enzyme-linked immunosorbent assay (ELISA) was used to quantify anti-inflammatory markers (TNF-α and IL-1β) 5 days after cells were exposed to PT-gliadin and/or anti-wheat gliadin IgY. RESULTS Among other conditions, anti-wheat gliadin IgY at a ratio of 1:3,000 (anti-gliadin IgY: PT-gliadin) significantly prevented gliadin toxicity on Caco-2 by maintaining intestinal integrity, inhibiting phenol red permeation, and inhibiting gliadin absorption and production of proinflammatory cytokines (TNF-α and IL-1β) as compared to PT-gliadin stimulated cultures (P < 0.05). CONCLUSION The anti-wheat gliadin IgY antibody produced in this study has proved to inhibit absorption of gliadin and gliadin-induced inflammatory response in Caco2 cell culture model of CD. Anti-gliadin IgY, therefore has potential to be used as an oral passive antibody therapy to treat CD.
Collapse
Affiliation(s)
- Naiyana Gujral
- 3142G Katz Group Centre for Pharmacy & Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 11361 - 87 Ave, Edmonton, AB, T6G 2E1, Canada.
| | - Ju Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin, Gyeonggi-Do, 449-728, Korea.
| | - Hoon H Sunwoo
- 3142G Katz Group Centre for Pharmacy & Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 11361 - 87 Ave, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
89
|
Abstract
Wheat-related disorders have become a growing area of clinical and scientific interest and can be categorized broadly as: autoimmune-mediated; allergic; and non-autoimmune/non-allergic conditions. Non-celiac gluten sensitivity (NCGS) and non-celiac wheat sensitivity (NCWS) present on this spectrum as disorders associated with adverse gastrointestinal and extra-intestinal manifestations following exposure to gluten and/or other wheat-related constituents. NCGS/NCWS is increasingly considered in patients with unexplained symptoms after the exclusions of celiac disease and wheat allergy. As objective diagnostic data and specific biomarkers are lacking, response to a gluten-free/wheat-free diet can confirm the presence of NCGS/NCWS. An association with irritable bowel syndrome has been detected, and the effects of other food components, such as fermentable oligosaccharides, disaccharides, monosaccharides, and polyols, may contribute. Our organization and synthesis of extant knowledge pertaining to wheat-related disorders may advance current practice and research efforts toward an improved understanding of NCGS/NCWS as an evolving clinical entity.
Collapse
Affiliation(s)
- Renée M Marchioni Beery
- Division of Gastroenterology and Hepatology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-1845, USA
| | | |
Collapse
|
90
|
Linee guida per la diagnosi di laboratorio e istologica della malattia celiaca. Revisione 2015. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s13631-015-0086-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
91
|
Bonavita R, Isticato R, Maurano F, Ricca E, Rossi M. Mucosal immunity induced by gliadin-presenting spores of Bacillus subtilis in HLA-DQ8-transgenic mice. Immunol Lett 2015; 165:84-9. [PMID: 25944582 DOI: 10.1016/j.imlet.2015.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/17/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
The induction of mucosal immunity requires efficient antigen delivery and adjuvant systems. Probiotic bacterial strains are considered to be very promising tools to address both of these needs. In particular, Bacillus subtilis spores are currently under investigation as a long-lived, protease-resistant adjuvant system for different antigens. Furthermore, a non-recombinant approach has been developed based on the stable adsorption of antigen on the spore surface. In the present study, we explored this strategy as a means of modulating the immune response to wheat gliadin, the triggering agent of celiac disease (CD), an enteropathy driven by inflammatory CD4(+) T cells. Gliadin adsorption was tested on untreated or autoclaved wild-type (wt) and mutant (cotH or cotE) spores. We found that gliadin was stably and maximally adsorbed by autoclaved wt spores. We then tested the immune properties of the spore-adsorbed gliadin in HLA-DQ8-transgenic mice, which express one of the two HLA heterodimers associated with CD. In vitro, spore-adsorbed gliadin was efficiently taken up by mouse dendritic cells (DCs). Interestingly, gliadin-pulsed DCs efficiently stimulated splenic CD4(+) T cells from mice immunised with spore-adsorbed gliadin. Nasal pre-dosing with spore-adsorbed gliadin failed to down-regulate the ongoing cellular response in gliadin-sensitised DQ8 mice. Notably, naïve mice inoculated intranasally with multiple doses of spore-adsorbed gliadin developed an intestinal antigen-specific CD4(+) T cell-mediated response. In conclusion, our data highlight the ability of spore-adsorbed gliadin to elicit a T-cell response in the gut that could be exploitable for developing immune strategies in CD.
Collapse
Affiliation(s)
| | | | | | - Ezio Ricca
- Department of Biology, Federico II University, Naples, Italy
| | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
92
|
van Bergen J, Mulder CJ, Mearin ML, Koning F. Local communication among mucosal immune cells in patients with celiac disease. Gastroenterology 2015; 148:1187-94. [PMID: 25623043 DOI: 10.1053/j.gastro.2015.01.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/14/2015] [Accepted: 01/20/2015] [Indexed: 12/16/2022]
Abstract
In patients with celiac disease, gluten consumption causes inflammation of the duodenum, and, to a lesser extent, the proximal jejunum. Immune-dominant gluten peptides are modified by the enzyme TG2, leading to their high-affinity binding to HLA-DQ2 or HLA-DQ8 molecules, present in people with a predisposition to celiac disease. Gluten peptide-loaded HLA-DQ2 or HLA-DQ8 molecules are recognized by highly conserved receptors on CD4(+) T cells in the lamina propria. B cells specific for TG2 and modified gluten peptides are also abundant in the lamina propria of patients with celiac disease. In the epithelium, interleukin-15 activates intraepithelial lymphocytes that promote destruction of epithelial cells. However, it is not clear how the immune responses in the lamina propria and the epithelium, separated by a basement membrane, are linked. We review the immune processes that occur in the lamina propria and their potential effects on epithelial pathology in celiac disease.
Collapse
Affiliation(s)
- Jeroen van Bergen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Mulder
- Department of Gastroenterology, Free University Medical Center, Amsterdam, The Netherlands
| | - M Luisa Mearin
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
93
|
Rossi E, Basso D, Zambon CF, Navaglia F, Greco E, Pelloso M, Artuso S, Padoan A, Pescarin M, Aita A, Bozzato D, Moz S, Cananzi M, Guariso G, Plebani M. TNFA Haplotype Genetic Testing Improves HLA in Estimating the Risk of Celiac Disease in Children. PLoS One 2015; 10:e0123244. [PMID: 25915602 PMCID: PMC4411089 DOI: 10.1371/journal.pone.0123244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/28/2015] [Indexed: 12/13/2022] Open
Abstract
Background TNF-α and IFN-γ play a role in the development of mucosal damage in celiac disease (CD). Polymorphisms of TNFA and IFNG genes, as well as of the TNFRSF1A gene, encoding the TNF-α receptor 1, might underlie different inter-individual disease susceptibility over a common HLA risk background. The aims of this study were to ascertain whether five SNPs in the TNFA promoter (-1031T>C,-857C>T,-376G>A,-308G>A,-238G>A), sequence variants of the TNFRSF1A gene and IFNG +874A>T polymorphism are associated with CD in a HLA independent manner. Methods 511 children (244 CD, 267 controls) were genotyped for HLA, TNFA and INFG (Real Time PCR). TNFRSF1A variants were studied (DHPLC and sequence). Results Only the rare TNFA-1031C (OR=0.65, 95% CI:0.44-0.95), -857T (OR=0.42, 95% CI:0.27-0.65), -376A (OR=2.25, 95% CI:1.12-4.51) and -308A (OR=4.76, 95% CI:3.12-7.26) alleles were significantly associated with CD. One TNFRSF1A variant was identified (c.625+10A>G, rs1800693), but not associated with CD. The CD-correlated TNFA SNPs resulted in six haplotypes. Two haplotypes were control-associated (CCGG and TTGG) and three were CD-associated (CCAG, TCGA and CCGA). The seventeen inferred haplotype combinations were grouped (A to E) based on their frequencies among CD. Binary logistic regression analysis documented a strong association between CD and HLA (OR for intermediate risk haplotypes=178; 95% CI:24-1317; OR for high risk haplotypes=2752; 95% CI:287-26387), but also an HLA-independent correlation between CD and TNFA haplotype combination groups. The CD risk for patients carrying an intermediate risk HLA haplotype could be sub-stratified by TNFA haplotype combinations. Conclusion TNFA promoter haplotypes associate with CD independently from HLA. We suggest that their evaluation might enhance the accuracy in estimating the CD genetic risk.
Collapse
Affiliation(s)
- Elisa Rossi
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Daniela Basso
- Department of Laboratory Medicine, University—Hospital of Padova, Padova, Italy
- * E-mail:
| | | | - Filippo Navaglia
- Department of Laboratory Medicine, University—Hospital of Padova, Padova, Italy
| | - Eliana Greco
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Michela Pelloso
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Serena Artuso
- Unit of Pediatric Gastroenterology, Department of Women and Children's Health, University-Hospital of Padova, Padova, Italy
| | - Andrea Padoan
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Matilde Pescarin
- Unit of Pediatric Gastroenterology, Department of Women and Children's Health, University-Hospital of Padova, Padova, Italy
| | - Ada Aita
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Dania Bozzato
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Stefania Moz
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| | - Mara Cananzi
- Unit of Pediatric Gastroenterology, Department of Women and Children's Health, University-Hospital of Padova, Padova, Italy
| | - Graziella Guariso
- Unit of Pediatric Gastroenterology, Department of Women and Children's Health, University-Hospital of Padova, Padova, Italy
| | - Mario Plebani
- Department of Medicine—DIMED, University of Padova, Padova, Italy
| |
Collapse
|
94
|
Abstract
BACKGROUND Celiac disease (CD) results from an alteration in the oral tolerance to dietary gluten. The response to gluten is normally tightly regulated and involves the secretion of TGF-β and IL-10 from different subtypes of regulatory T cells (Tregs). Interestingly, in addition to proinflammatory cytokines, the inflamed CD mucosa also contains high levels of T cell-derived IL-10 compared with treated CD patients or normal donors. Furthermore, most studies describe an increase in the number of Foxp3+ Tregs in the small intestinal mucosa in CD patients compared to controls. This paradoxical condition suggests that regulatory mechanisms might operate to counterbalance the abnormal gliadin-triggered immune activation in untreated mucosa. Indeed, addition of exogenous IL-10 to mucosal cultures from treated CD patients can suppress gliadin-induced T cell activation. Considering the central role of adaptive immunity in CD, the development of strategies to stimulate these mechanisms is a primary goal of efforts to restore gluten tolerance. Key Messages: Different immunomodulatory strategies have been explored. NexVax2, a desensitizing vaccine that uses three dominant gluten peptides administered subcutaneously to induce a tolerogenic response in CD patients, is under development. Alternatively, the potential of substituted, cyclic or dimeric peptide analogues as blockers to prevent HLA from binding to the immunodominant gliadin epitopes has been demonstrated in vitro. In line with these results, we recently found that modified (transamidated) gliadins influenced the immune response in intestinal biopsy samples from CD patients with overt disease by drastically reducing the production of IFN-γ. Notably, in a mouse model, transamidated gliadins reverted the phenotype of the gliadin-inducible immune response from an inflammatory phenotype to an anti-inflammatory phenotype. CONCLUSIONS Various approaches are currently under investigation to recover gluten tolerance based on the use of both modified and native antigen molecules. More specific studies are now required to test the efficacy of such strategies for preventing CD.
Collapse
Affiliation(s)
- Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy
| |
Collapse
|
95
|
Castillo NE, Theethira TG, Leffler DA. The present and the future in the diagnosis and management of celiac disease. Gastroenterol Rep (Oxf) 2015; 3:3-11. [PMID: 25326000 PMCID: PMC4324867 DOI: 10.1093/gastro/gou065] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/26/2014] [Indexed: 12/19/2022] Open
Abstract
Celiac disease is an autoimmune enteropathy caused by gluten in genetically predisposed individuals. In celiac disease, adaptive and innate immune activation results in intestinal damage and a wide range of clinical manifestations. In the past, celiac disease was thought to result in signs and symptoms solely related to the gastrointestinal tract. Now, more than half of the adult population presents with extra-intestinal manifestations that can also be expected to improve on a gluten-free diet. For this reason, it is recommended that physicians have a low threshold of suspicion for celiac disease. Current knowledge of the immune pathogenesis of this autoimmune disease has served as a catalyst for the development of novel diagnostic tools and therapeutics. Over the years, highly sensitive and specific serological assays, in addition to genetic markers, have been found to target specific steps in the cascade pathway of celiac disease. Also the advent of the gluten challenge has enabled experts to design diagnostic algorithms and monitor clinical responses in clinical trials. The gluten challenge has provided substantial benefit in the advance of novel therapeutics as an adjuvant treatment to the gluten free diet. Generally, a strict gluten-free diet is highly burdensome to patients and can be limited in its efficacy. Alternative therapies-including gluten modification, modulation of intestinal permeability and immune response-could be central to the future treatment of celiac disease.
Collapse
Affiliation(s)
- Natalia E Castillo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Daniel A Leffler
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
96
|
Abstract
Interleukin-15 (IL-15) exerts many biological functions essential for the maintenance and function of multiple cell types. Although its expression is tightly regulated, IL-15 upregulation has been reported in many organ-specific autoimmune disorders. In celiac disease, an intestinal inflammatory disorder driven by gluten exposure, the upregulation of IL-15 expression in the intestinal mucosa has become a hallmark of the disease. Interestingly, because it is overexpressed both in the gut epithelium and in the lamina propria, IL-15 acts on distinct cell types and impacts distinct immune components and pathways to disrupt intestinal immune homeostasis. In this article, we review our current knowledge of the multifaceted roles of IL-15 with regard to the main immunological processes involved in the pathogenesis of celiac disease.
Collapse
Affiliation(s)
- Valérie Abadie
- Sainte-Justine Hospital Research Center, Department of Microbiology and Immunology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | | |
Collapse
|
97
|
Abadie V, Jabri B. Immunopathology of Celiac Disease. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00080-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
98
|
Vincentini O, Maialetti F, Gonnelli E, Silano M. Gliadin-dependent cytokine production in a bidimensional cellular model of celiac intestinal mucosa. Clin Exp Med 2014; 15:447-54. [PMID: 25447031 DOI: 10.1007/s10238-014-0325-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/20/2014] [Indexed: 12/19/2022]
Abstract
The downstream cascade of the inflammatory response to gliadin in celiac intestinal mucosa encompasses the early activation of the innate immunity that triggers the adaptive response. Therefore, the in vitro study of the pathogenic mechanism of celiac disease (CD) on enterocytes alone or mucosal T lymphocytes alone does not fully consider all the aspects of gliadin-dependent inflammation. Although the in vitro culture of specimens of intestinal mucosa obtained from celiac patients is the gold standard for the study of CD, this technique presents several technical challenges and the bioptic specimens are not easily available. So, in this paper, we described the gliadin-dependent cytokine production in a bidimensional cellular system, which is able to mimic both the innate and the adaptive steps of the mucosal immune response of CD. In the upper compartment, the intestinal epithelial cells are grown on a filter, and in the lower compartment, the mononuclear cells isolated from peripheral blood of celiac patients are cultured. Cells were apically exposed to the toxic gliadin peptide p31-43 for 3 h and then with the immunodominant gliadin fragment pα-9 for 21 h. The incubation with gliadin peptides resulted in increased levels of IL-15, INF-γ, IL-6, tumor necrosis factor (TNF)-α, IL-1β, and CCL 2, 3 and 4 in the basal supernatants, with respect to cells exposed to medium alone. The p31-43-driven epithelial priming of mucosal response consists of transglutaminase (TG2)-mediated deamidation of the immunostimulatory gliadin peptides, as demonstrated by the inhibition of pα-9 activity, when the system is exposed to blocking anti-TG2 antibody.
Collapse
Affiliation(s)
- Olimpia Vincentini
- Unit of Human Nutrition and Health, Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità - Italian National Institute of Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Maialetti
- Unit of Human Nutrition and Health, Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità - Italian National Institute of Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Elena Gonnelli
- Unit of Human Nutrition and Health, Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità - Italian National Institute of Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marco Silano
- Unit of Human Nutrition and Health, Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità - Italian National Institute of Health, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
99
|
Nikoukar L, Nabavizadeh F, Mohamadi S, Moslehi A, Hassanzadeh G, Nahrevanian H, Agah S. Protective effect of ghrelin in a rat model of celiac disease. ACTA ACUST UNITED AC 2014; 101:438-47. [DOI: 10.1556/aphysiol.101.2014.4.5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
100
|
De Palma G, Vida C, Santacruz A, De Castro NM, De la Fuente M, Sanz Y. Impaired responses to gliadin and gut microbes of immune cells from mice with altered stress-related behavior and premature immune senescence. J Neuroimmunol 2014; 276:47-57. [PMID: 25176132 DOI: 10.1016/j.jneuroim.2014.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/03/2014] [Accepted: 08/06/2014] [Indexed: 01/22/2023]
Abstract
Stress is associated with impaired communication between the nervous and immune systems leading to immunosenescence and increased disease risk. We investigated whether leukocytes from mice with altered stress-related behavior and premature immunosenescence, as well as from chronologically aged mice differently responded ex vivo to celiac disease (CD) triggers (gliadin) and intestinal bacteria by ELISA and flow cytometry and differed in microbiota composition. We found that altered stress-related behavior and premature immunosenescence led to alterations in T lymphocytes and cytokine release of immune cells basally and in response to peptic fragments of gliadin and commensal and pathogenic bacteria, possibly increasing susceptibility to CD in adulthood.
Collapse
Affiliation(s)
- G De Palma
- Microbial Ecology, Nutrition & Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - C Vida
- Department of Physiology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - A Santacruz
- Microbial Ecology, Nutrition & Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - N M De Castro
- Department of Physiology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - M De la Fuente
- Department of Physiology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Y Sanz
- Microbial Ecology, Nutrition & Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|