1
|
Fiske KL, Brigleb PH, Sanchez LM, Hinterleitner R, Taylor GM, Dermody TS. Strain-specific differences in reovirus infection of murine macrophages segregate with polymorphisms in viral outer-capsid protein σ3. J Virol 2024; 98:e0114724. [PMID: 39431846 PMCID: PMC11575339 DOI: 10.1128/jvi.01147-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/22/2024] [Indexed: 10/22/2024] Open
Abstract
Mammalian orthoreovirus (reovirus) strains type 1 Lang (T1L) and type 3 Dearing-RV (T3D-RV) infect the intestine in mice but differ in the induction of inflammatory responses. T1L infection is associated with the blockade of oral immunological tolerance to newly introduced dietary antigens, whereas T3D-RV is not. T1L infection leads to an increase in infiltrating phagocytes, including macrophages, in gut-associated lymphoid tissues that are not observed in T3D-RV infection. However, the function of macrophages in reovirus intestinal infection is unknown. Using cells sorted from infected intestinal tissue and primary cultures of bone-marrow-derived macrophages (BMDMs), we discovered that T1L infects macrophages more efficiently than T3D-RV. Analysis of T1L × T3D-RV reassortant viruses revealed that the viral S4 gene segment, which encodes outer-capsid protein σ3, is responsible for strain-specific differences in infection of BMDMs. Differences in the binding of T1L and T3D-RV to BMDMs also segregated with the σ3-encoding S4 gene. Paired immunoglobulin-like receptor B (PirB), which serves as a receptor for reovirus, is expressed on macrophages and engages σ3. We found that PirB-specific antibody blocks T1L binding to BMDMs and that T1L binding to PirB-/- BMDMs is significantly diminished. Collectively, our data suggest that reovirus T1L infection of macrophages is dependent on engagement of PirB by viral outer-capsid protein σ3. These findings raise the possibility that macrophages function in the innate immune response to reovirus infection that blocks immunological tolerance to new food antigens.IMPORTANCEMammalian orthoreovirus (reovirus) infects humans throughout their lifespan and has been linked to celiac disease (CeD). CeD is caused by a loss of oral immunological tolerance (LOT) to dietary gluten and leads to intestinal inflammation following gluten ingestion, which worsens with prolonged exposure and can cause malnutrition. There are limited treatment options for CeD. While there are genetic risk factors associated with the illness, triggers for disease onset are not completely understood. Enteric viruses, including reovirus, have been linked to CeD induction. We found that a reovirus strain associated with oral immunological tolerance blockade infects macrophages by virtue of its capacity to bind macrophage receptor PirB. These data contribute to an understanding of the innate immune response elicited by reovirus, which may shed light on how viruses trigger LOT and inform the development of CeD vaccines and therapeutic agents.
Collapse
Affiliation(s)
- Kay L. Fiske
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pamela H. Brigleb
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Luzmariel Medina Sanchez
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Reinhard Hinterleitner
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gwen M. Taylor
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
Renga G, Pariano M, D'Onofrio F, Pieraccini G, Di Serio C, Villella VR, Abbate C, Puccetti M, Giovagnoli S, Stincardini C, Bellet MM, Ricci M, Costantini C, Oikonomou V, Romani L. The immune and microbial homeostasis determines the Candida-mast cells cross-talk in celiac disease. Life Sci Alliance 2024; 7:e202302441. [PMID: 38719750 PMCID: PMC11079604 DOI: 10.26508/lsa.202302441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Fiorella D'Onofrio
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Claudia Di Serio
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Abbate
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | | | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Abadie V, Han AS, Jabri B, Sollid LM. New Insights on Genes, Gluten, and Immunopathogenesis of Celiac Disease. Gastroenterology 2024; 167:4-22. [PMID: 38670280 PMCID: PMC11283582 DOI: 10.1053/j.gastro.2024.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois.
| | - Arnold S Han
- Columbia Center for Translational Immunology, Columbia University, New York, New York; Department of Microbiology and Immunology, Columbia University, New York, New York; Department of Medicine, Digestive and Liver Diseases, Columbia University, New York, New York
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois; Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
4
|
Medina Sanchez L, Siller M, Zeng Y, Brigleb PH, Sangani KA, Soto AS, Engl C, Laughlin CR, Rana M, Van Der Kraak L, Pandey SP, Bender MJ, Fitzgerald B, Hedden L, Fiske K, Taylor GM, Wright AP, Mehta ID, Rahman SA, Galipeau HJ, Mullett SJ, Gelhaus SL, Watkins SC, Bercik P, Nice TJ, Jabri B, Meisel M, Das J, Dermody TS, Verdú EF, Hinterleitner R. The gut protist Tritrichomonas arnold restrains virus-mediated loss of oral tolerance by modulating dietary antigen-presenting dendritic cells. Immunity 2023; 56:1862-1875.e9. [PMID: 37478853 PMCID: PMC10529081 DOI: 10.1016/j.immuni.2023.06.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 03/29/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023]
Abstract
Loss of oral tolerance (LOT) to gluten, driven by dendritic cell (DC) priming of gluten-specific T helper 1 (Th1) cell immune responses, is a hallmark of celiac disease (CeD) and can be triggered by enteric viral infections. Whether certain commensals can moderate virus-mediated LOT remains elusive. Here, using a mouse model of virus-mediated LOT, we discovered that the gut-colonizing protist Tritrichomonas (T.) arnold promotes oral tolerance and protects against reovirus- and murine norovirus-mediated LOT, independent of the microbiota. Protection was not attributable to antiviral host responses or T. arnold-mediated innate type 2 immunity. Mechanistically, T. arnold directly restrained the proinflammatory program in dietary antigen-presenting DCs, subsequently limiting Th1 and promoting regulatory T cell responses. Finally, analysis of fecal microbiomes showed that T. arnold-related Parabasalid strains are underrepresented in human CeD patients. Altogether, these findings will motivate further exploration of oral-tolerance-promoting protists in CeD and other immune-mediated food sensitivities.
Collapse
Affiliation(s)
- Luzmariel Medina Sanchez
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magdalena Siller
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yanlin Zeng
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; School of Medicine, Tsinghua University, Beijing, China
| | - Pamela H Brigleb
- Graduate Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kishan A Sangani
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Ariadna S Soto
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Clarisse Engl
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Colin R Laughlin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohit Rana
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Van Der Kraak
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Surya P Pandey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mackenzie J Bender
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Britney Fitzgerald
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lee Hedden
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kay Fiske
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gwen M Taylor
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Austin P Wright
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Isha D Mehta
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Syed A Rahman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Health Sciences Mass Spectrometry Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stacy L Gelhaus
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Health Sciences Mass Spectrometry Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Terence S Dermody
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elena F Verdú
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Crocco M, Calvi A, Canzoneri F, Malerba F, Zampatti N, Chiaro A, Arrigo S, Gandullia P, Proietti S, Bonassi S. The Influence of SARS-CoV-2 Pandemic on the Diagnosis of Celiac Disease and Clinical Practice in Pediatric Gastroenterology. Nutrients 2023; 15:nu15030559. [PMID: 36771266 PMCID: PMC9920531 DOI: 10.3390/nu15030559] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/14/2023] [Indexed: 01/24/2023] Open
Abstract
Celiac disease (CD) has a high prevalence but remains largely underdiagnosed. Although extensive studies have confirmed that children with CD do not have an increased risk of severe COVID-19, public health regulations associated with the SARS-CoV-2 pandemic may have exacerbated this problem. The aim of this study was to assess the effect of SARS-CoV-2 on the number of new-onset CD cases. Additionally, the role of SARS-CoV-2 in autoimmune diseases and its influence on clinical practice in pediatric gastroenterology were briefly reviewed. We described the data from the hospital electronic registry of new-onset CD, during the COVID-19 pandemic and 2 years before. A total of 423 children were diagnosed with CD between March 2018 and February 2022: 228 in the 2-year pre-COVID-19 period and 195 during the pandemic. The number of patients during the COVID-19 pandemic was 14.5% lower than in the previous years. The quarterly comparison of CD diagnoses showed a reduction in all quarters. A reduction in diagnoses during the lockdown and in the following months was evident and not compensated thereafter. This is the first study to evaluate the impact of SARS-CoV-2 on the diagnosis of CD in children. Further studies are necessary to improve the system of biopsy-sparing diagnosis and to evaluate the effect of the diagnostic delay. Special attention should be given to the implementation of telemedicine services.
Collapse
Affiliation(s)
- Marco Crocco
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, 16132 Genova, Italy
- Correspondence:
| | - Angela Calvi
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Francesca Canzoneri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, 16132 Genova, Italy
| | - Federica Malerba
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, 16132 Genova, Italy
| | - Noemi Zampatti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, 16132 Genova, Italy
| | - Andrea Chiaro
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Serena Arrigo
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Paolo Gandullia
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Stefania Proietti
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Stefano Bonassi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| |
Collapse
|
6
|
Zoghi S, Abbasi A, Heravi FS, Somi MH, Nikniaz Z, Moaddab SY, Ebrahimzadeh Leylabadlo H. The gut microbiota and celiac disease: Pathophysiology, current perspective and new therapeutic approaches. Crit Rev Food Sci Nutr 2022; 64:2176-2196. [PMID: 36154539 DOI: 10.1080/10408398.2022.2121262] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Celiac disease (CD) as a chronic gluten-sensitive intestinal condition, mainly affects genetically susceptible hosts. The primary determinants of CD have been identified as environmental and genetic variables. The development of CD is significantly influenced by environmental factors, including the gut microbiome. Therefore, gut microbiome re-programming-based therapies using probiotics, prebiotics, postbiotics, gluten-free diet, and fecal microbiota transplantation have shown promising results in the modification of the gut microbiome. Due to the importance and paucity of information regarding the CD pathophysiology, in this review, we have covered the association between CD development and gut microbiota, the effects of infectious agents, particularly the recent Covid-19 infection in CD patients, and the efficacy of potential therapeutic approaches in the CD have been discussed. Hence, scientific literature indicates that the diverse biological functions of the gut microbiota against immunomodulatory responses have made microbiome-based therapy an alternative therapeutic paradigm to ameliorate the symptoms of CD and quality of life. However, the exact potential of microbiota-based techniques that aims to quantitatively and qualitatively alter the gut microbiota to be used in the treatment and ameliorate the symptoms of CD will be determined with further research in the future.
Collapse
Affiliation(s)
- Sevda Zoghi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Yaghoub Moaddab
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
7
|
Seroprevalence of Anti-tTg-IgA among Symptomized Celiac Disease Patients and Their Correlation with Rotavirus Infection. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6972624. [PMID: 36193310 PMCID: PMC9526599 DOI: 10.1155/2022/6972624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022]
Abstract
Celiac disease (CD) is a chronic inflammatory disorder in the intestinal tract as a response to the use of gluten in genetically predisposed individuals. It is a worldwide problem, with a high prevalence rate in North America. This is a descriptive cross-sectional study involving 1090 samples collected from different hospitals of Rawalpindi and Islamabad, Pakistan, from January 2019 to December 2019. In this study, 1090 blood samples screened for seroprevalence of anti-tTG antibodies in CD symptomatic patients via ELISA (enzyme-linked immunosorbent assay). 1090 fecal samples from the same CD patients were collected and tested for the presence of rotavirus (RV) via ELISA and RT-PCR. Of the 1090 patients tested for seroprevalence of anti-tTG antibodies, 112/1090 (10.3%) were found to be positive. Of the 112 anti-tTG-positive patients, 78/112 (70%) were positive for RV via ELISA and 74/112 (66.1%) were RV positive via RT-PCR. A statistically significant association was reported between rotavirus infection and celiac disease (p˂0.05). Anti-tTG antibodies were higher in age group 6 (12-18 years) patients (18.4%) and at minimum in age group 3 (1-3 years) patients (4.8%). However, there was a statistically insignificant relationship between group age and CD prevalence (p > 0.05). The highest CD prevalence was noted during winter season (19.6%) and the lowest (3.0%) during fall/autumn. Our study findings demonstrate that Pakistan has a high prevalence of CD compared to other studies. Further studies in the fields of environmental risk factors and treatment with more advanced serological and histopathological studies are needed in the future.
Collapse
|
8
|
Brigleb PH, Kouame E, Fiske KL, Taylor GM, Urbanek K, Medina Sanchez L, Hinterleitner R, Jabri B, Dermody TS. NK cells contribute to reovirus-induced IFN responses and loss of tolerance to dietary antigen. JCI Insight 2022; 7:159823. [PMID: 35993365 PMCID: PMC9462493 DOI: 10.1172/jci.insight.159823] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Celiac disease is an immune-mediated intestinal disorder that results from loss of oral tolerance (LOT) to dietary gluten. Reovirus elicits inflammatory Th1 cells and suppresses Treg responses to dietary antigen in a strain-dependent manner. Strain type 1 Lang (T1L) breaks oral tolerance, while strain type 3 Dearing reassortant virus (T3D-RV) does not. We discovered that intestinal infection by T1L in mice leads to the recruitment and activation of NK cells in mesenteric lymph nodes (MLNs) in a type I IFN-dependent manner. Once activated following infection, NK cells produce type II IFN and contribute to IFN-stimulated gene expression in the MLNs, which in turn induces inflammatory DC and T cell responses. Immune depletion of NK cells impairs T1L-induced LOT to newly introduced food antigen. These studies indicate that NK cells modulate the response to dietary antigen in the presence of a viral infection.
Collapse
Affiliation(s)
- Pamela H. Brigleb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elaine Kouame
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| | - Kay L. Fiske
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pediatrics and
| | - Gwen M. Taylor
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pediatrics and
| | - Kelly Urbanek
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pediatrics and
| | - Luzmariel Medina Sanchez
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA.,Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Terence S. Dermody
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pediatrics and
| |
Collapse
|
9
|
Celdir MG, Jansson-Knodell CL, Hujoel IA, Prokop LJ, Wang Z, Murad MH, Murray JA. Latitude and Celiac Disease Prevalence: A Meta-Analysis and Meta-Regression. Clin Gastroenterol Hepatol 2022; 20:e1231-e1239. [PMID: 33007509 DOI: 10.1016/j.cgh.2020.09.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/15/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The latitudinal gradient effect is described for several autoimmune diseases including celiac disease in the United States. However, the association between latitude and global celiac disease prevalence is unknown. We aimed to explore the association between latitude and serology-based celiac disease prevalence through meta-analysis. METHODS We searched MEDLINE, Embase, Cochrane, and Scopus databases from their beginning through June 29, 2018, to identify screening studies that targeted a general population sample, used serology-based screening tests, and provided a clear location from which we could assign a latitude. Studies were excluded if sampling was based on symptoms, risk factors, or referral. Study selection and data extraction were performed by independent reviewers. The association measures between latitude and prevalence of serology-based celiac disease were evaluated with random-effects meta-analyses and meta-regression. RESULTS Of the identified 4667 unique citations, 128 studies were included, with 155 prevalence estimates representing 40 countries. Celiac disease was more prevalent at the higher latitudes of 51° to 60° (relative risk [RR], 1.62; 95% CI, 1.09-2.38) and 61° to 70° (RR, 2.30; 95% CI, 1.36-3.89) compared with the 41° to 50° reference level. No statistically significant difference was observed at lower latitudes. When latitude was treated as continuous, we found a statistically significant association between CD prevalence and latitude overall in the world (RR, 1.03, 95% CI, 1.01-1.05) and a subregional analysis of Europe (RR, 1.05; 95% CI, 1.02-1.07) and North America (RR, 1.1; 95% CI, 1.0-1.2). CONCLUSIONS In this comprehensive review of screening studies, we found that a higher latitude was associated with greater serology-based celiac disease prevalence.
Collapse
Affiliation(s)
- Melis G Celdir
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Isabel A Hujoel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Zhen Wang
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota
| | - M Hassan Murad
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
10
|
Saha BK, Saha S, Bonnier A, Saha BN. Association between idiopathic pulmonary hemosiderosis and celiac disease in pediatric patients: A scoping review of the literature over the past 50 years. Pediatr Pulmonol 2022; 57:1127-1144. [PMID: 35088581 DOI: 10.1002/ppul.25847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Idiopathic pulmonary hemosiderosis (IPH) is a rare cause of diffuse alveolar hemorrhage, the mechanism of which is currently unknown. Nearly one-third of pediatric patients with IPH test positive for Celiac disease (CD) serology. Several hypothetical mechanisms have been proposed to unify the coexistence of these two entities, also referred to as Lane-Hamilton syndrome (LHS). METHOD This manuscript is a scoping review of the medical literature. Medline, Embase, and PubMed Central databases were searched between 1971 and 2021 with appropriate search words to identify all cases of pediatric LHS. RESULTS A total of 20 manuscripts with 23 pediatric patients with LHS were identified. The mean age was 11 years, and 13/23 (56.5%) of the children were boys. Hemoptysis was present in 57% of patients during diagnosis. Bronchoscopy with bronchoalveolar lavage demonstrating hemosiderin laden macrophages was the primary mode of diagnostic confirmation. Only three patients underwent lung biopsy. Any significant GI symptom was reported in a minority of patients (22%). Iron deficiency anemia on presentation was described in 83% of children. The majority of patients were malnourished. Serology for CD was positive in all patients, as was the histopathologic analysis of the small bowel biopsy. No patients had any other autoantibody positivity. The introduction of gluten free diet (GFD) was associated with a positive response in 20/23 patients. CONCLUSION All pediatric patients with IPH should undergo screening for CD. Low serum ferritin in patients with IPH could be suggestive of coexisting CD. Strict GFD should be tried as the initial therapy.
Collapse
Affiliation(s)
- Biplab K Saha
- Department of Pulmonary and Critical Care Medicine, Ozarks Medical Center, West Plains, Missouri, USA
| | - Santu Saha
- Department of Internal Medicine, Bangladesh Medical College, Dhaka, Bangladesh
| | - Alyssa Bonnier
- Department of Critical Care Nursing, Goldfarb School of Nursing, Barnes Jewish College, St. Louis, Missouri, USA
| | | |
Collapse
|
11
|
Saha BK, Datar P, Aiman A, Bonnier A, Saha S, Milman NT. Comparative Analysis of Adult Patients With Idiopathic Pulmonary Hemosiderosis and Lane-Hamilton Syndrome: A Systematic Review of the Literature in the Period 1971-2022. Cureus 2022; 14:e23482. [PMID: 35475077 PMCID: PMC9035284 DOI: 10.7759/cureus.23482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 12/16/2022] Open
Abstract
Idiopathic pulmonary hemosiderosis (IPH) causes diffuse alveolar hemorrhage (DAH) by a yet unknown mechanism. The coexistence of IPH and celiac disease (CD), also known as Lane-Hamilton syndrome (LHS), has been reported in both pediatric and adult patients. The objective of this study was to compare demographics, clinical and radiologic findings, treatment, and outcomes between adult patients with IPH and LHS. This is a systematic review of the literature. Multiple databases were searched using appropriate formulas to identify relevant articles. A total of 60 studies reporting 65 patients were included in the review. Forty-nine of these patients had IPH and 16 had LHS. The prevalence of anti-CD antibodies among tested patients was 13/22 (59%). The symptom onset and diagnosis of IPH occurred earlier in patients with LHS. The median delay in diagnosis was the same between the two groups (52 weeks). The classic triad was more likely to be present in patients with LHS. Only 20% of patients in the LHS cohort had any significant gastrointestinal (GI) symptoms at the time of IPH diagnosis. A gluten-free diet alone was effective in the majority of patients. Fewer patients in the LHS cohort received systemic corticosteroid than the IPH cohort. The recurrence and mortality in patients with LHS appear to be less than in the IPH cohort. The prevalence of CD is 25% in adult patients with IPH. Patients with LHS may have a milder course than patients without CD. Serologic testing for CD should be performed in all patients diagnosed with IPH.
Collapse
|
12
|
Abstract
Celiac disease is a chronic, immune-mediated enteropathy driven by dietary gluten found in genetically susceptible hosts. It has a worldwide distribution, is one of the most common autoimmune disorders globally, and is the only autoimmune condition for which the trigger is known. Despite advances in characterizing mechanisms of disease, gaps in understanding of celiac disease pathogenesis remain. A "frontier" concept is considering what moves an HLA-DQ2 or DQ8-positive individual from asymptomatic gluten tolerance to celiac disease manifestation. In this arena, environmental triggers, including age at the time of initial gluten exposure, the occurrence of usual childhood viral infections, and microbiome alterations have emerged as key events in triggering the symptomatic disease. Pathologists play a major role in frontier aspects of celiac disease. This includes the discovery that duodenal mucosal histology in follow-up biopsies does not correlate with ongoing patient symptoms, antitissue transglutaminase antibody titers and diet adherence in celiac disease patients. Further, in light of recent evidence that the detection of monoclonal T-cell populations in formalin-fixed biopsies is not specific for type II refractory celiac disease, pathologists should resist performing such analyses until common causes of "apparent" refractoriness are excluded. The promise of therapies in celiac disease has led to clinical trials targeting many steps in the inflammatory cascade, which depend upon a pathologist's confirmation of the initial diagnosis and evaluation of responses to therapies. As pathologists continue to be active participants in celiac disease research, partnering with other stakeholders, we will continue to impact this important autoimmune disease.
Collapse
Affiliation(s)
- Natalie Patel
- El Camino Pathology Medical Group, Mountain View, CA
| | - Marie E Robert
- Department of Pathology and Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
13
|
Voisine J, Abadie V. Interplay Between Gluten, HLA, Innate and Adaptive Immunity Orchestrates the Development of Coeliac Disease. Front Immunol 2021; 12:674313. [PMID: 34149709 PMCID: PMC8206552 DOI: 10.3389/fimmu.2021.674313] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Several environmental, genetic, and immune factors create a "perfect storm" for the development of coeliac disease: the antigen gluten, the strong association of coeliac disease with HLA, the deamidation of gluten peptides by the enzyme transglutaminase 2 (TG2) generating peptides that bind strongly to the predisposing HLA-DQ2 or HLA-DQ8 molecules, and the ensuing unrestrained T cell response. T cell immunity is at the center of the disease contributing to the inflammatory process through the loss of tolerance to gluten and the differentiation of HLA-DQ2 or HLA-DQ8-restricted anti-gluten inflammatory CD4+ T cells secreting pro-inflammatory cytokines and to the killing of intestinal epithelial cells by cytotoxic intraepithelial CD8+ lymphocytes. However, recent studies emphasize that the individual contribution of each of these cell subsets is not sufficient and that interactions between these different populations of T cells and the simultaneous activation of innate and adaptive immune pathways in distinct gut compartments are required to promote disease immunopathology. In this review, we will discuss how tissue destruction in the context of coeliac disease results from the complex interactions between gluten, HLA molecules, TG2, and multiple innate and adaptive immune components.
Collapse
Affiliation(s)
- Jordan Voisine
- Department of Medicine, The University of Chicago, Chicago, IL, United States.,Committee on Immunology, The University of Chicago, Chicago, IL, United States
| | - Valérie Abadie
- Department of Medicine, The University of Chicago, Chicago, IL, United States.,Section of Gastroenterology, Nutrition and Hepatology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Gasmi A, Tippairote T, Mujawdiya PK, Peana M, Menzel A, Dadar M, Benahmed AG, Bjørklund G. The microbiota-mediated dietary and nutritional interventions for COVID-19. Clin Immunol 2021; 226:108725. [PMID: 33845194 PMCID: PMC8032598 DOI: 10.1016/j.clim.2021.108725] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Worldwide, scientists are looking for specific treatment for COVID-19. Apart from the antiviral approach, the interventions to support healthy immune responses to the virus are feasible through diet, nutrition, and lifestyle approaches. This narrative review explores the recent studies on dietary, nutritional, and lifestyle interventions that influence the microbiota-mediated immunomodulatory effects against viral infections. Cumulative studies reported that the airway microbiota and SARS-CoV-2 leverage each other and determine the pathogen-microbiota-host responses. Cigarette smoking can disrupt microbiota abundance. The composition and diversification of intestinal microbiota influence the airway microbiota and the innate and adaptive immunity, which require supports from the balance of macro- and micronutrients from the diet. Colorful vegetables supplied fermentable prebiotics and anti-inflammatory, antioxidant phytonutrients. Fermented foods and beverages support intestinal microbiota. In sensitive individuals, the avoidance of the high immunoreactive food antigens contributes to antiviral immunity. This review suggests associations between airway and intestinal microbiota, antiviral host immunity, and the influences of dietary, nutritional, and lifestyle interventions to prevent the clinical course toward severe COVID-19.
Collapse
Affiliation(s)
- Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Torsak Tippairote
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine, Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok, Thailand; Thailand Institute for Functional Medicine, Bangkok, Thailand
| | | | | | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| |
Collapse
|
15
|
Akobeng AK, Singh P, Kumar M, Al Khodor S. Role of the gut microbiota in the pathogenesis of coeliac disease and potential therapeutic implications. Eur J Nutr 2020; 59:3369-3390. [PMID: 32651763 PMCID: PMC7669811 DOI: 10.1007/s00394-020-02324-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Although genetic predisposition and exposure to dietary gluten are considered necessary triggers for the development of coeliac disease, alterations in the gut microbial composition may also contribute towards the pathogenesis of coeliac disease. This review aims to provide an overview of the available data on the potential mechanisms through which the gut microbiota plays a role in the causation of coeliac disease and to discuss the potential therapeutic strategies that could diminish the consequences of microbial dysbiosis. METHOD A search of the literature was performed using the PubMed, Embase, and JSTOR databases; relevant articles were included. RESULTS Recent studies in patients with coeliac disease have reported an increase in the relative amounts of gram negative bacterial genera such as Bacteroides, Prevotella, and Escherichia, and reduced amounts of protective anti-inflammatory bacteria such as Bifidobacteria and Lactobacilli. Dysbiotic microbiota may lead to a dysregulated immune response that may contribute to the pathogenesis of coeliac disease. In infancy, antibiotic use and certain infant feeding practices may lead to alterations in the developing gut microbiota to influence the immune maturation process and predispose to coeliac disease. CONCLUSION The induction of the intestinal immune system and gluten intolerance may be influenced by the relative abundance of certain microbiota. Factors such as infant feeding practices, diet, antibiotics, and infections, may be involved in the development of coeliac disease due to their influence on gut microbial composition. The efficacy of potential modulators of the gut microbiota such as probiotics, prebiotics, and fecal microbial transplant as adjunctive treatments to gluten-free diet in coeliac disease is unproven and requires further investigation.
Collapse
Affiliation(s)
- Anthony K Akobeng
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Cornell University, Doha, Qatar
| | - Parul Singh
- Research Department, Sidra Medicine, Doha, Qatar
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
16
|
Ramírez-Sánchez AD, Tan IL, Gonera-de Jong B, Visschedijk MC, Jonkers I, Withoff S. Molecular Biomarkers for Celiac Disease: Past, Present and Future. Int J Mol Sci 2020; 21:E8528. [PMID: 33198309 PMCID: PMC7697360 DOI: 10.3390/ijms21228528] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
Celiac disease (CeD) is a complex immune-mediated disorder that is triggered by dietary gluten in genetically predisposed individuals. CeD is characterized by inflammation and villous atrophy of the small intestine, which can lead to gastrointestinal complaints, malnutrition, and malignancies. Currently, diagnosis of CeD relies on serology (antibodies against transglutaminase and endomysium) and small-intestinal biopsies. Since small-intestinal biopsies require invasive upper-endoscopy, and serology cannot predict CeD in an early stage or be used for monitoring disease after initiation of a gluten-free diet, the search for non-invasive biomarkers is ongoing. Here, we summarize current and up-and-coming non-invasive biomarkers that may be able to predict, diagnose, and monitor the progression of CeD. We further discuss how current and emerging techniques, such as (single-cell) transcriptomics and genomics, can be used to uncover the pathophysiology of CeD and identify non-invasive biomarkers.
Collapse
Affiliation(s)
- Aarón D. Ramírez-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (A.D.R.-S.); (I.L.T.); (I.J.)
| | - Ineke L. Tan
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (A.D.R.-S.); (I.L.T.); (I.J.)
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| | - B.C. Gonera-de Jong
- Department of Pediatrics, Wilhelmina Hospital Assen, 9401 RK Assen, The Netherlands;
| | - Marijn C. Visschedijk
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| | - Iris Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (A.D.R.-S.); (I.L.T.); (I.J.)
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (A.D.R.-S.); (I.L.T.); (I.J.)
| |
Collapse
|
17
|
Reed AW, Dabrowski A, Joseph S, Paranjape SM, Karwowski C. Lane-Hamilton Syndrome: An Illustration of Extraintestinal Manifestations of Celiac Disease. Clin Pediatr (Phila) 2020; 59:1195-1198. [PMID: 32686478 DOI: 10.1177/0009922820941210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Anna W Reed
- Johns Hopkins Children's Center, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
18
|
Dunne MR, Byrne G, Chirdo FG, Feighery C. Coeliac Disease Pathogenesis: The Uncertainties of a Well-Known Immune Mediated Disorder. Front Immunol 2020; 11:1374. [PMID: 32733456 PMCID: PMC7360848 DOI: 10.3389/fimmu.2020.01374] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Coeliac disease is a common small bowel enteropathy arising in genetically predisposed individuals and caused by ingestion of gluten in the diet. Great advances have been made in understanding the role of the adaptive immune system in response to gluten peptides. Despite detailed knowledge of these adaptive immune mechanisms, the complete series of pathogenic events responsible for development of the tissue lesion remains less certain. This review contributes to the field by discussing additional mechanisms which may also contribute to pathogenesis. These include the production of cytokines such as interleukin-15 by intestinal epithelial cells and local antigen presenting cells as a pivotal event in the disease process. A subset of unconventional T cells called gamma/delta T cells are also persistently expanded in the coeliac disease (CD) small intestinal epithelium and recent analysis has shown that these cells contribute to pathogenic inflammation. Other unconventional T cell subsets may play a local immunoregulatory role and require further study. It has also been suggested that, in addition to activation of pathogenic T helper cells by gluten peptides, other peptides may directly interact with the intestinal mucosa, further contributing to the disease process. We also discuss how myofibroblasts, a major source of tissue transglutaminase and metalloproteases, may play a key role in intestinal tissue remodeling. Contribution of each of these factors to pathogenesis is discussed to enhance our view of this complex disorder and to contribute to a wider understanding of chronic immune-mediated disease.
Collapse
Affiliation(s)
- Margaret R. Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Greg Byrne
- School of Biological & Health Sciences, Technological University, Dublin, Ireland
| | - Fernando G. Chirdo
- Instituto de Estudios Inmunologicos y Fisiopatologicos - IIFP (UNLP-CONICET), National University of La Plata, La Plata, Argentina
| | - Conleth Feighery
- Department of Immunology, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
19
|
Jauregi-Miguel A, Santin I, Garcia-Etxebarria K, Olazagoitia-Garmendia A, Romero-Garmendia I, Sebastian-delaCruz M, Irastorza I, Castellanos-Rubio A, Bilbao JR. MAGI2 Gene Region and Celiac Disease. Front Nutr 2019; 6:187. [PMID: 31921880 PMCID: PMC6930898 DOI: 10.3389/fnut.2019.00187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Celiac disease (CD) patients present a loss of intestinal barrier function due to structural alterations in the tight junction (TJ) network, the most apical unions between epithelial cells. The association of TJ-related gene variants points to an implication of this network in disease susceptibility. This work aims to characterize the functional implication of TJ-related, disease-associated loci in CD pathogenesis. We performed an association study of 8 TJ-related gene variants in a cohort of 270 CD and 91 non-CD controls. The expression level of transcripts located in the associated SNP region was analyzed by RT-PCR in several human tissues and in duodenal biopsies of celiac patients and non-CD controls. (si)RNA-driven silencing combined with gliadin in the Caco2 intestinal cell line was used to analyze the implication of transcripts from the associated region in the regulation of TJ genes. We replicated the association of rs6962966*A variant [p = 0.0029; OR = 1.88 (95%1.24–2.87)], located in an intron of TJ-related MAGI2 coding gene and upstream of RP4-587D13.2 transcript, bioinformatically classified as a long non-coding RNA (lncRNA). The expression of both genes is correlated and constitutively downregulated in CD intestine. Silencing of lncRNA decreases the levels of MAGI2 protein. At the same time, silencing of MAGI2 affects the expression of several TJ-related genes. The associated region is functionally altered in disease, probably affecting CD-related TJ genes.
Collapse
Affiliation(s)
- Amaia Jauregi-Miguel
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain
| | - Izortze Santin
- Department of Biochemistry and Molecular Biology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain.,CIBER in Diabetes and Associated Metabolic Diseases, Madrid, Spain
| | - Koldo Garcia-Etxebarria
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain
| | - Ane Olazagoitia-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain
| | - Irati Romero-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain
| | - Maialen Sebastian-delaCruz
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain
| | - Iñaki Irastorza
- Department of Pediatrics, Biocruces-Bizkaia Health Research Institute, Cruces University Hospital, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Barakaldo, Spain
| | | | - Ainara Castellanos-Rubio
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Jose Ramón Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces-Bizkaia Health Research Institute, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), Leioa, Spain.,CIBER in Diabetes and Associated Metabolic Diseases, Madrid, Spain
| |
Collapse
|
20
|
Shariati A, Aslani HR, Shayesteh MR, Taghipour A, Nasser A, Safari H, Alizade-Sani M, Dehghan A, Azimi T. Are Viruses and Parasites Linked to Celiac Disease? A Question that Still has no Definite Answer. Curr Pharm Biotechnol 2019; 20:1181-1193. [PMID: 31456516 DOI: 10.2174/1389201020666190828124924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/01/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
Celiac Disease (CD) is a complex autoimmune enteropathy of the small intestine that commonly
occurs in genetically predisposed individuals due to intake of gluten and related proteins. Gluten
consumption, duration of breast-feeding, various infections, especially frequent intestinal infections,
vaccinations and use of antibiotics can be linked to CD. It is predicted that it affects 1% of the
global population and its incidence rate is increasing. Most of the people with the HLA-DQ2 or HLADQ8
are at a higher risk of developing this disease. The link between infections and autoimmune diseases
has been very much considered in recent years. In several studies, we explained that pathogenic
and non-pathogenic microorganisms might have multiple roles in initiation, exacerbation, and development
of Irritable Bowel Syndrome (IBS) and Inflammatory Bowel Disease (IBD). In various studies,
the relationship between infections caused by viruses, such as Epstein-Barr Virus (EBV), Rotavirus,
Hepatitis C (HCV), Hepatitis B virus (HBV), Cytomegalovirus (CMV), and Influenza virus, and parasites
including Giardia spp. and Toxoplasma gondii with CD has been raised. However, increasing evidence
proposes that some of these microorganisms, especially helminths, can also have protective and
even therapeutic roles in the CD process. Therefore, in order to determine the role of microorganisms
in the process of this disease, we attempted to summarize the evidence suggesting the role of viral and
parasitic agents in pathogenesis of CD.
Collapse
Affiliation(s)
- Aref Shariati
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid R. Aslani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad R.H. Shayesteh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Taghipour
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ahmad Nasser
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Safari
- Health Promotion Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Alizade-Sani
- Food Safety and Hygiene Division, Environmental Health Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Dehghan
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Taher Azimi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Autoimmune and Allergic Disorders are More Common in People With Celiac Disease or on a Gluten-free Diet in the United States. J Clin Gastroenterol 2019; 53:e416-e423. [PMID: 30045167 DOI: 10.1097/mcg.0000000000001100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
GOALS We analyzed demographics, lifestyle patterns, and clinical characteristics of people with celiac disease (CD) and people without CD avoiding gluten (PWAG) to better understand associations with medical conditions and consumer behavior. BACKGROUND Clinical significance of CD and gluten avoidance in the general population is incompletely understood. Recently, a high incidence of CD in adolescents with susceptibility genotypes, similar to other autoimmune or allergic disorders, and regional differences in consumer practices of gluten avoidance were reported. METHODS Among 22,277 participants in the National Health and Nutrition Examination Survey 2009-2014, we identified persons with CD by testing CD serology or by both a health care provider diagnosis and adherence to a gluten-free diet. Similarly, PWAG were defined as adherent to a gluten-free diet without a CD diagnosis. Consumer behavior and characteristics of both groups, CD and PWAG were compared with those without these conditions, using survey-weighted generalized logistic regression. RESULTS Participants with CD considered nutrition very important when grocery shopping and tended to have more constipation and thyroid disease. PWAG tended to spend more money on groceries, purchase organic foods, and check food labels more frequently during grocery shopping. They also reported having more food allergies, asthma, and thyroid disease. CONCLUSIONS Our study confirms that CD and PWAG share comorbidities of autoimmune nature. PWAG had more autoimmune/allergy-related disorders that may be associated with non-celiac gluten sensitivity a self-justifiable reason to be on the diet.
Collapse
|
22
|
Uusitalo U, Andren Aronsson C, Liu X, Kurppa K, Yang J, Liu E, Skidmore J, Winkler C, Rewers MJ, Hagopian WA, She JX, Toppari J, Ziegler AG, Akolkar B, Norris JM, Virtanen SM, Krischer JP, Agardh D. Early Probiotic Supplementation and the Risk of Celiac Disease in Children at Genetic Risk. Nutrients 2019; 11:nu11081790. [PMID: 31382440 PMCID: PMC6722940 DOI: 10.3390/nu11081790] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/20/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
Probiotics are linked to positive regulatory effects on the immune system. The aim of the study was to examine the association between the exposure of probiotics via dietary supplements or via infant formula by the age of 1 year and the development of celiac disease autoimmunity (CDA) and celiac disease among a cohort of 6520 genetically susceptible children. Use of probiotics during the first year of life was reported by 1460 children. Time-to-event analysis was used to examine the associations. Overall exposure of probiotics during the first year of life was not associated with either CDA (n = 1212) (HR 1.15; 95%CI 0.99, 1.35; p = 0.07) or celiac disease (n = 455) (HR 1.11; 95%CI 0.86, 1.43; p = 0.43) when adjusting for known risk factors. Intake of probiotic dietary supplements, however, was associated with a slightly increased risk of CDA (HR 1.18; 95%CI 1.00, 1.40; p = 0.043) compared to children who did not get probiotics. It was concluded that the overall exposure of probiotics during the first year of life was not associated with CDA or celiac disease in children at genetic risk.
Collapse
Grants
- U01 DK063821 NIDDK NIH HHS
- UC4 DK063863 NIDDK NIH HHS
- U01 DK63829, U01 DK63861, U01 DK63821, U01 DK63865, U01 DK63863, U01 DK63836, U01 DK63790, UC4 DK63829, UC4 DK63861, UC4 DK63821, UC4 DK63865, UC4 DK63863, UC4 DK63836, UC4 DK95300, UC4 DK100238, UC4 DK106955, UC4 DK112243, UC4 DK117483 NIDDK NIH HHS
- UL1 TR000064 NIH HHS
- UC4 DK117483 NIDDK NIH HHS
- UC4 DK112243 NIDDK NIH HHS
- UL1 TR001082 NIH HHS
- U01 DK063863 NIDDK NIH HHS
- UC4 DK106955 NIDDK NIH HHS
- HHSN267200700014C NIDDK NIH HHS
Collapse
Affiliation(s)
- Ulla Uusitalo
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Carin Andren Aronsson
- The Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| | - Xiang Liu
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kalle Kurppa
- Tampere Center for Child Health Research, University of Tampere, 33014 Tampere, Finland
- The University Consortium of Seinäjoki, 60320 Seinäjoki, Finland
| | - Jimin Yang
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Edwin Liu
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado Denver, Aurora, CO 80045, USA
| | | | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Zentrum München, 85764 Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 80804 Munich, Germany
- Forschergruppe Diabetes e.V., Helmholtz Zentrum München, 85764 Munich-Neuherberg, Germany
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | - Jin-Xiong She
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20014 Turku, Finland
- Department of Pediatrics, Turku University Hospital, 20521 Turku, Finland
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, 85764 Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 80804 Munich, Germany
- Forschergruppe Diabetes e.V., Helmholtz Zentrum München, 85764 Munich-Neuherberg, Germany
| | - Beena Akolkar
- NIDDK, National Institute of Health, Bethesda, MD 20892, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, CO 80045, USA
| | - Suvi M Virtanen
- Tampere Center for Child Health Research, University of Tampere, 33014 Tampere, Finland
- Unit of Nutrition, National Institute for Health and Welfare, 00271 Helsinki, Finland
- Tampere University Hospital, and the Science Center of Pirkanmaa Hospital District, 33520 Tampere, Finland
| | - Jeffrey P Krischer
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Daniel Agardh
- The Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| |
Collapse
|
23
|
Abstract
Purpose of Review Mammalian orthoreovirus (reovirus) is a powerful tool for studying viral replication and pathogenesis. Most reovirus infections are subclinical, however recent work has catapulted reovirus into the clinical spotlight. Recent Findings Owing to its capacity to kill cancer cells more efficiently than normal cells, reovirus is under development as a therapeutic for a variety of cancers. New efforts have focused on genetically engineering reovirus to increase its oncolytic capacity, and determining how reovirus potentiates immunotherapy. Other recent studies highlight a potential role for reovirus in celiac disease (CeD). Using mouse models of CeD, reovirus caused loss of oral tolerance to dietary antigens, opening the possibility that reovirus could trigger CeD in humans. Summary We will focus on new developments in reovirus oncolysis and studies suggesting a role for reovirus as a trigger for celiac disease (CeD) that make reovirus a potential friend and foe to human health.
Collapse
|
24
|
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
|
25
|
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
|
26
|
Moerkens R, Mooiweer J, Withoff S, Wijmenga C. Celiac disease-on-chip: Modeling a multifactorial disease in vitro. United European Gastroenterol J 2019; 7:467-476. [PMID: 31065364 PMCID: PMC6488795 DOI: 10.1177/2050640619836057] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022] Open
Abstract
Conventional model systems cannot fully recapitulate the multifactorial character of complex diseases like celiac disease (CeD), a common chronic intestinal disorder in which many different genetic risk factors interact with environmental factors such as dietary gluten. However, by combining recently developed human induced pluripotent stem cell (hiPSC) technology and organ-on-chip technology, in vitro intestine-on-chip systems can now be developed that integrate the genetic background of complex diseases, the different interacting cell types involved in disease pathology, and the modulating environmental factors such as gluten and the gut microbiome. The hiPSCs that are the basis of these systems can be generated from both diseased and healthy individuals, which means they can be stratified based on their load of genetic risk factors. A CeD-on-chip model system has great potential to improve our understanding of disease etiology and accelerate the development of novel treatments and preventive therapies in CeD and other complex diseases.
Collapse
Affiliation(s)
- Renée Moerkens
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joram Mooiweer
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,K.G. Jebsen Coeliac Disease Research Center, Department of Immunology, University of Oslo, Norway
| |
Collapse
|
27
|
Abstract
Coeliac disease (CD) is an immune-mediated disorder triggered by the ingestion of gluten in genetically susceptible individuals. However, only a small proportion of subjects harbouring CD-related genetic risk develop the disease. Among the environmental factors that may influence CD risk, pre- and perinatal factors, delivery methods, parental lifestyle, infant feeding practices, seasonality, dietary factors, drug use, childhood infections and variability in gut microbiota are those most widely studied regarding the risk to develop CD. Although for many of these external factors the exact mechanism of action is unknown, most of them are thought to act by disrupting the intestinal barrier, facilitating contact between potential antigens and the immune system effector cells. Management of CD is relatively easy in patients with a definite diagnosis and requires a strict, lifelong, gluten-free diet. Better knowledge of environmental exposures apart from gluten can facilitate understanding of the pathogenesis of the disorder and the wide heterogeneity of its clinical spectrum. The purpose of this review is to discuss current knowledge on environmental CD risk factors, as well as possible interaction between them, on the grounds of the reliable scientific evidence available. Key messages The risk of developing CD is influenced not only by gluten ingestion but also by a number of environmental factors including childhood infections and variability in gut microbiota, pre- and perinatal factors, infant feeding practices, delivery methods, parental lifestyle, seasonality, dietary factors and drug use, acting mainly by disrupting intestinal permeability. Better knowledge of exposure to these factors can facilitate their identification, and subsequent elimination, in the individual patient.
Collapse
Affiliation(s)
- Giovanni Mario Pes
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy
| | - Stefano Bibbò
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy
| | - Maria Pina Dore
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy.,b Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
28
|
Abstract
Finely tuned mechanisms enable the gastrointestinal tract to break down dietary components into nutrients without mounting, in the majority of cases, a dysregulated immune or functional host response. However, adverse reactions to food have been steadily increasing, and evidence suggests that this process is environmental. Adverse food reactions can be divided according to their underlying pathophysiology into food intolerances, when, for instance, there is deficiency of a host enzyme required to digest the food component, and food sensitivities, when immune mechanisms are involved. In this Review, we discuss the clinical and experimental evidence for enteric infections and/or alterations in the gut microbiota in inciting food sensitivity. We focus on mechanisms by which microorganisms might provide direct pro-inflammatory signals to the host promoting breakdown of oral tolerance to food antigens or indirect pathways that involve the metabolism of protein antigens and other dietary components by gut microorganisms. Better understanding of these mechanisms will help in the development of preventive and therapeutic strategies for food sensitivities.
Collapse
|
29
|
Gupta S, Kaushal A, Kumar A, Kumar D. Recent advances in biosensors for diagnosis of celiac disease: A review. Biotechnol Bioeng 2018; 116:444-451. [PMID: 30516838 DOI: 10.1002/bit.26856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 09/13/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022]
Abstract
Celiac disease (CD) is an intestinal issue activated by the inappropriate immune reaction towards gluten protein of wheat, rye, barley, oats, and autoantigen, tissue transglutaminase. Regardless of the accessibility of immunochemical conventions for research facility analysis of CD, there is as yet a need of speedier, less expensive, and simpler devices for diagnosing CD. This review concentrates on progresses in biosensors for diagnosing CD in perspective of the scaled down hardware, multianalyte discovery and low sample volume necessity. Various recently developed biosensors in this field are presented.
Collapse
Affiliation(s)
- Shagun Gupta
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| | - Ankur Kaushal
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India.,Department of Molecular Biosensor lab, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Ashok Kumar
- Department of Molecular Biosensor lab, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Dinesh Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| |
Collapse
|
30
|
Bouziat R, Biering SB, Kouame E, Sangani KA, Kang S, Ernest JD, Varma M, Brown JJ, Urbanek K, Dermody TS, Ng A, Hinterleitner R, Hwang S, Jabri B. Murine Norovirus Infection Induces T H1 Inflammatory Responses to Dietary Antigens. Cell Host Microbe 2018; 24:677-688.e5. [PMID: 30392830 PMCID: PMC6326098 DOI: 10.1016/j.chom.2018.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/03/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
Intestinal reovirus infection can trigger T helper 1 (TH1) immunity to dietary antigen, raising the question of whether other viruses can have a similar impact. Here we show that the acute CW3 strain of murine norovirus, but not the persistent CR6 strain, induces TH1 immunity to dietary antigen. This property of CW3 is dependent on its major capsid protein, a virulence determinant. Transcriptional profiling of mesenteric lymph nodes following infection reveals an immunopathological signature that does not segregate with protective immunity but with loss of oral tolerance, in which interferon regulatory factor 1 is critical. These data show that viral capacity to trigger specific inflammatory pathways at sites where T cell responses to dietary antigens take place interferes with the development of tolerance to an oral antigen. Collectively, these data provide a foundation for the development of therapeutic strategies to prevent TH1-mediated complex immune disorders triggered by viral infections.
Collapse
Affiliation(s)
- Romain Bouziat
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Scott B Biering
- Committee on Microbiology, University of Chicago, Chicago, IL, USA
| | - Elaine Kouame
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Kishan A Sangani
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Soowon Kang
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, 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
| | - Judy J Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly Urbanek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Terence S Dermody
- 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
| | - 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
| | - Reinhard Hinterleitner
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Seungmin Hwang
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Committee on Microbiology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA.
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
31
|
Affiliation(s)
- Judy J. Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Bana Jabri
- Department of Medicine and Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Olivares M, Benítez-Páez A, de Palma G, Capilla A, Nova E, Castillejo G, Varea V, Marcos A, Garrote JA, Polanco I, Donat E, Ribes-Koninckx C, Calvo C, Ortigosa L, Palau F, Sanz Y. Increased prevalence of pathogenic bacteria in the gut microbiota of infants at risk of developing celiac disease: The PROFICEL study. Gut Microbes 2018; 9:551-558. [PMID: 29672211 PMCID: PMC6287676 DOI: 10.1080/19490976.2018.1451276] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy involving genetic and environmental factors, whose interaction influences disease risk. The intestinal microbiota, including viruses and bacteria, could play a role in the pathological process leading to gluten intolerance. In this study, we investigated the prevalence of pathogens in the intestinal microbiota of infants at familial risk of developing CD. We included 127 full-term newborns with at least one first-degree relative with CD. Infants were classified according to milk-feeding practice (breastfeeding or formula feeding) and HLA-DQ genotype (low, intermediate or high genetic risk). The prevalence of pathogenic bacteria and viruses was assessed in the faeces of the infants at 7 days, 1 month and 4 months of age. The prevalence of Clostridium perfringens was higher in formula-fed infants than in breast-fed over the study period, and that of C. difficile at 4 months. Among breastfed infants, a higher prevalence of enterotoxigenic E. coli (ETEC) was found in infants with the highest genetic risk compared either to those with a low or intermediate risk. Among formula-fed infants, a higher prevalence of ETEC was also found in infants with a high genetic risk compared to those of intermediate risk. Our results show that specific factors, such as formula feeding and the HLA-DQ2 genotype, previously linked to a higher risk of developing CD, influence the presence of pathogenic bacteria differently in the intestinal microbiota in early life. Further studies are warranted to establish whether these associations are related to CD onset later in life.
Collapse
Affiliation(s)
- Marta Olivares
- Microbial Ecology, Nutrition & Health Research Unit. Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain,CONTACT Marta Olivares IATA-CSIC, C/Catedrático Agustín Escardino, 7. 46980, Paterna, Valencia, Spain
| | - Alfonso Benítez-Páez
- Microbial Ecology, Nutrition & Health Research Unit. Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Giada de Palma
- Microbial Ecology, Nutrition & Health Research Unit. Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Amalia Capilla
- Genetics and Molecular Medicine Unit. Institute of Biomedicine of Valencia, Spanish National Research Council (IBV-CSIC), Valencia, Spain
| | - Esther Nova
- Institute of Food Science, Technology and Nutrition, Spanish National Research Council (ICTAN-CSIC), Madrid, Spain
| | - Gemma Castillejo
- Universitary Hospital Sant Joan of Reus, URV, IISPV, Tarragona, Spain
| | - Vicente Varea
- Gastroenterología, Nutrición y Hepatología Pediátrica, Hospital Universitario Sant Joan de Deu, and Instituto de Gastroeneterología y Nutrición Pediátrica de Barcelona, Barcelona, Spain
| | - Ascensión Marcos
- Institute of Food Science, Technology and Nutrition, Spanish National Research Council (ICTAN-CSIC), Madrid, Spain
| | - José Antonio Garrote
- Research Unit and Paediatric Service, Hospital Clínico Universitario, Valladolid, Spain
| | - Isabel Polanco
- Servicio de Gastroenterología y Nutrición Pediátrica, Hospital Universitario La Paz, Madrid, Spain
| | - Ester Donat
- Unidad de Gastroenterología, Hospital Infantil Universitario La Fe, Valencia, Spain
| | | | - Carmen Calvo
- Research Unit and Paediatric Service, Hospital Clínico Universitario, Valladolid, Spain
| | - Luis Ortigosa
- Unidad de Gastroenterologia, Hepatología y Nutrición Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain
| | - Francesc Palau
- Genetics and Molecular Medicine Unit. Institute of Biomedicine of Valencia, Spanish National Research Council (IBV-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition & Health Research Unit. Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain,Yolanda Sanz IATA-CSIC, C/Catedrático Agustín Escardino, 7. 46980, Paterna, Valencia, Spain
| |
Collapse
|
33
|
Amil Dias J. Celiac Disease: What Do We Know in 2017? GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2017; 24:275-278. [PMID: 29255768 PMCID: PMC5731182 DOI: 10.1159/000479881] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/31/2017] [Indexed: 12/20/2022]
Abstract
Celiac disease is one of the most prevalent digestive conditions. Diagnosis requires that strict criteria are used so that a life-long gluten-free diet may be correctly prescribed. Although genetic susceptibility has been known for a long time, there have been elusive environmental factors that lead to the occurrence of clinical disease. Many studies have addressed the identification of environmental modifiers, and different lines of research have been tried with variable success and even contradictory results. Infections and age of gluten introduction into the diet in the first few months of life have been evaluated, but a firm relationship could not be established. A recent paper addresses a fascinating hypothesis that could explain how some infectious agents might modulate the immune system and modify response to dietary antigens. Subsequently, animal models with genetic susceptibility were tested, and, indeed, there was abnormal response to gluten. These observations still do not provide final answers about the pathophysiology of celiac disease but certainly lead to progress in the knowledge of gluten sensitization and the role of some environmental factors.
Collapse
Affiliation(s)
- Jorge Amil Dias
- Pediatra e Gastrenterologista Pediátrico, Unidade de Gastrenterologia Pediátrica, Centro Hospitalar S. João, Porto, Portugal
| |
Collapse
|
34
|
Inulin-Type Fructans Application in Gluten-Free Products: Functionality and Health Benefits. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-3-319-54528-8_2-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
35
|
Alaedini A, Lebwohl B, Wormser GP, Green PH, Ludvigsson JF. Borrelia infection and risk of celiac disease. BMC Med 2017; 15:169. [PMID: 28911326 PMCID: PMC5599869 DOI: 10.1186/s12916-017-0926-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Environmental factors, including infectious agents, are speculated to play a role in the rising prevalence and the geographic distribution of celiac disease, an autoimmune disorder. In the USA and Sweden where the regional variation in the frequency of celiac disease has been studied, a similarity with the geographic distribution of Lyme disease, an emerging multisystemic infection caused by Borrelia burgdorferi spirochetes, has been found, thus raising the possibility of a link. We aimed to determine if infection with Borrelia contributes to an increased risk of celiac disease. METHODS Biopsy reports from all of Sweden's pathology departments were used to identify 15,769 individuals with celiac disease. Through linkage to the nationwide Patient Register, we compared the rate of earlier occurrence of Lyme disease in the patients with celiac disease to that in 78,331 matched controls. To further assess the temporal relationship between Borrelia infection and celiac disease, we also examined the risk of subsequent Lyme disease in patients with a diagnosis of celiac disease. RESULTS Twenty-five individuals (0.16%) with celiac disease had a prior diagnosis of Lyme disease, whereas 79 (0.5%) had a subsequent diagnosis of Lyme disease. A modest association between Lyme disease and celiac disease was seen both before (odds ratio, 1.61; 95% confidence interval (CI), 1.06-2.47) and after the diagnosis of celiac disease (hazard ratio, 1.82; 95% CI, 1.40-2.35), with the risk of disease being highest in the first year of follow-up. CONCLUSIONS Only a minor fraction of the celiac disease patient population had a prior diagnosis of Lyme disease. The similar association between Lyme disease and celiac disease both before and after the diagnosis of celiac disease is strongly suggestive of surveillance bias as a likely contributor. Taken together, the data indicate that Borrelia infection is not a substantive risk factor in the development of celiac disease.
Collapse
Affiliation(s)
- Armin Alaedini
- Department of Medicine, Columbia University Medical Center, New York, NY, USA.
- Celiac Disease Center, Columbia University Medical Center, New York, NY, USA.
- Institute of Human Nutrition, Columbia University Medical Center, New York, NY, USA.
| | - Benjamin Lebwohl
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Celiac Disease Center, Columbia University Medical Center, New York, NY, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gary P Wormser
- Division of Infectious Diseases, Department of Medicine, New York Medical College, Valhalla, NY, USA
| | - Peter H Green
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Celiac Disease Center, Columbia University Medical Center, New York, NY, USA
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Department of Pediatrics, Örebro University Hospital, Örebro, Sweden.
- Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
36
|
Lerner A, Arleevskaya M, Schmiedl A, Matthias T. Microbes and Viruses Are Bugging the Gut in Celiac Disease. Are They Friends or Foes? Front Microbiol 2017; 8:1392. [PMID: 28824555 PMCID: PMC5539691 DOI: 10.3389/fmicb.2017.01392] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/10/2017] [Indexed: 12/17/2022] Open
Abstract
The links between microorganisms/viruses and autoimmunity are complex and multidirectional. A huge number of studies demonstrated the triggering impact of microbes and viruses as the major environmental factors on the autoimmune and inflammatory diseases. However, growing evidences suggest that infectious agents can also play a protective role or even abrogate these processes. This protective crosstalk between microbes/viruses and us might represent a mutual beneficial equilibrium relationship between two cohabiting ecosystems. The protective pathways might involve post-translational modification of proteins, decreased intestinal permeability, Th1 to Th2 immune shift, induction of apoptosis, auto-aggressive cells relocation from the target organ, immunosuppressive extracellular vesicles and down regulation of auto-reactive cells by the microbial derived proteins. Our analysis demonstrates that the interaction of the microorganisms/viruses and celiac disease (CD) is always a set of multidirectional processes. A deeper inquiry into the CD interplay with Herpes viruses and Helicobacter pylori demonstrates that the role of these infections, suggested to be potential CD protectors, is not as controversial as for the other infectious agents. The outcome of these interactions might be due to a balance between these multidirectional processes.
Collapse
Affiliation(s)
- Aaron Lerner
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of TechnologyHaifa, Israel
- Department of Research, AESKU.KIPP InstituteWendelsheim, Germany
| | - Marina Arleevskaya
- Central Research Laboratory, Kazan State Medical Academy KazanKazan, Russia
| | - Andreas Schmiedl
- Department of Research, AESKU.KIPP InstituteWendelsheim, Germany
| | - Torsten Matthias
- Department of Research, AESKU.KIPP InstituteWendelsheim, Germany
| |
Collapse
|
37
|
Campagna G, Pesce M, Tatangelo R, Rizzuto A, La Fratta I, Grilli A. The progression of coeliac disease: its neurological and psychiatric implications. Nutr Res Rev 2017; 30:25-35. [PMID: 27976606 DOI: 10.1017/s0954422416000214] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of the paper is to show the various neurological and psychiatric symptoms in coeliac disease (CD). CD is a T cell-mediated, tissue-specific autoimmune disease which affects genetically susceptible individuals after dietary exposure to proline- and glutamine-rich proteins contained in certain cereal grains. Genetics, environmental factors and different immune systems, together with the presence of auto-antigens, are taken into account when identifying the pathogenesis of CD. CD pathogenesis is related to immune dysregulation, which involves the gastrointestinal system, and the extra-intestinal systems such as the nervous system, whose neurological symptoms are evidenced in CD patients. A gluten-free diet (GFD) could avoid cerebellar ataxia, epilepsy, neuropathies, migraine and mild cognitive impairment. Furthermore, untreated CD patients have more symptoms and psychiatric co-morbidities than those treated with a GFD. Common psychiatric symptoms in untreated CD adult patients include depression, apathy, anxiety, and irritability and schizophrenia is also common in untreated CD. Several studies show improvement in psychiatric symptoms after the start of a GFD. The present review discusses the state of the art regarding neurological and psychiatric complications in CD and highlights the evidence supporting a role for GFD in reducing neurological and psychiatric complications.
Collapse
Affiliation(s)
- Giovanna Campagna
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| | - Mirko Pesce
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| | - Raffaella Tatangelo
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| | - Alessia Rizzuto
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| | - Irene La Fratta
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| | - Alfredo Grilli
- Medicine and Health Science School,Università "G. d'Annunzio",Via dei Vestini,31,66100 Chieti CH,Italy
| |
Collapse
|
38
|
Jansen MAE, Beth SA, van den Heuvel D, Kiefte-de Jong JC, Raat H, Jaddoe VWV, van Zelm MC, Moll HA. Ethnic differences in coeliac disease autoimmunity in childhood: the Generation R Study. Arch Dis Child 2017; 102:529-534. [PMID: 28052882 DOI: 10.1136/archdischild-2016-311343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 12/05/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The aim was to identify whether ethnic differences in coeliac disease autoimmunity (CDA) in children at 6 years of age exist, and when present, to evaluate how these differences may be explained by sociodemographic and environmental factors. DESIGN This study was embedded within a multi-ethnic population-based prospective cohort study. SETTING AND PATIENTS 4442 six-year-old children born between 2002 and 2006 were included. Information on ethnicity, environmental and lifestyle characteristics was assessed by questionnaires. Ethnicity was categorised into Western (Dutch, European, Indonesian, American, Oceanian) and non-Western (Turkish, Moroccan, Cape Verdean, Antillean, Surinamese). Serum transglutaminase type 2 antibody (TG2A) levels were measured with fluorescence enzyme immunoassay. Serum IgG levels against cytomegalovirus (CMV) were measured by ELISA. MAIN OUTCOME MEASURES TG2A positivity was defined as TG2A ≥7 U/mL, strong TG2A positivity as TG2A ≥10 upper limit normal (70 U/mL). RESULTS Of 4442 children, 60 (1.4%) children were TG2A positive, of whom 31 were strong positive. 66% of children were Western, 33% non-Western. Western ethnicity, high socioeconomic position and daycare attendance were positively associated with strong TG2A positivity (odds ratio (OR) 6.85 (1.62 to 28.8) p<0.01, OR 3.70 (1.40 to 9.82) p<0.01, OR 3.90 (1.38 to 11.0) p=0.01 resp.), whereas CMV seropositivity was inversely related to strong TG2A positivity (OR 0.32 (0.12 to 0.84) p=0.02). Together, these factors explained up to 47% (-67 to -17; p=0.02) of the ethnic differences in TG2A positivity between Western and non-Western children. CONCLUSIONS Ethnic differences in children with CDA are present in childhood. Socioeconomic position, daycare attendance and CMV seropositivity partly explained these differences, which may serve as targets for prevention strategies for CDA.
Collapse
Affiliation(s)
- Michelle A E Jansen
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Sytske A Beth
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Diana van den Heuvel
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jessica C Kiefte-de Jong
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Global Public Health, Leiden University College, The Hague, The Netherlands
| | - Hein Raat
- Department of Public Health, University Medical Center, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Henriette A Moll
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
39
|
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: 335] [Impact Index Per Article: 41.9] [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
|
40
|
García-Álvarez M, Alcoceba M, López-Parra M, Puig N, Antón A, Balanzategui A, Prieto-Conde I, Jiménez C, Sarasquete ME, Chillón MC, Gutiérrez ML, Corral R, Alonso JM, Queizán JA, Vidán J, Pardal E, Peñarrubia MJ, Bastida JM, García-Sanz R, Marín L, González M. HLA specificities are associated with prognosis in IGHV-mutated CLL-like high-count monoclonal B cell lymphocytosis. PLoS One 2017; 12:e0172978. [PMID: 28249016 PMCID: PMC5332061 DOI: 10.1371/journal.pone.0172978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/12/2017] [Indexed: 12/18/2022] Open
Abstract
Introduction Molecular alterations leading progression of asymptomatic CLL-like high-count monoclonal B lymphocytosis (hiMBL) to chronic lymphocytic leukemia (CLL) remain poorly understood. Recently, genome-wide association studies have found 6p21.3, where the human leukocyte antigen (HLA) system is coded, to be a susceptibility risk region for CLL. Previous studies have produced discrepant results regarding the association between HLA and CLL development and outcome, but no studies have been performed on hiMBL. Aims We evaluated the role of HLA class I (-A, -B and -C) and class II (-DRB1 and -DQB1) in hiMBL/CLL susceptibility, hiMBL progression to CLL, and treatment requirement in a large series of 263 patients diagnosed in our center with hiMBL (n = 156) or Binet A CLL (n = 107). Results No consistent association between HLA specificities and hiMBL or CLL susceptibility was found. With a median follow-up of 7.7 years, 48/156 hiMBLs (33%) evolved to asymptomatic CLLs, while 16 hiMBLs (10%) and 44 CLLs (41%) required treatment. No HLA specificities were found to be significantly associated with hiMBL progression or treatment in the whole cohort. However, within antigen-experienced immunoglobulin heavy-chain (IGHV)-mutated hiMBLs, which represents the highest proportion of hiMBL cases (81%), the presence of HLA-DQB1*03 showed a trend to a higher risk of progression to CLL (60% vs. 26%, P = 0.062). Moreover, HLA-DQB1*02 specificity was associated with a lesser requirement for 15-year treatment (10% vs. 36%, P = 0.012). Conclusion In conclusion, our results suggest a role for HLA in IGHV-mutated hiMBL prognosis, and are consistent with the growing evidence of the influence of 6p21 on predisposition to CLL. Larger non-biased series are required to enable definitive conclusions to be drawn.
Collapse
Affiliation(s)
- María García-Álvarez
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Miguel Alcoceba
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
- CIBERONC, Madrid, Spain
| | - Miriam López-Parra
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Noemí Puig
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Alicia Antón
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Ana Balanzategui
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Isabel Prieto-Conde
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Cristina Jiménez
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - María E. Sarasquete
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - M. Carmen Chillón
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - María Laura Gutiérrez
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL (University of Salamanca), Salamanca, Spain
| | - Rocío Corral
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - José María Alonso
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
| | - José Antonio Queizán
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
| | - Julia Vidán
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
| | - Emilia Pardal
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
| | - María Jesús Peñarrubia
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
| | - José M. Bastida
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
| | - Ramón García-Sanz
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
- * E-mail:
| | - Luis Marín
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - Marcos González
- Department of Hematology, University Hospital of Salamanca (HUS-IBSAL), Salamanca, Spain
- Cooperative Working Group on Lymphomas and Lymphoproliferative Disorders of the Castilla y León Society of Hematology and Hemotherapy (SCLHH), Castilla y León, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
41
|
O. Temajo N, Howard N. The divergence between the virus and cellular oxidative stress as separate environmental agents that trigger autoimmunity originates from their different procedural mechanisms of activating the same molecular entity: the transcription factor NF-kappa B. AIMS ALLERGY AND IMMUNOLOGY 2017. [DOI: 10.3934/allergy.2017.2.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
42
|
Kanduc D, Shoenfeld Y. From HBV to HPV: Designing vaccines for extensive and intensive vaccination campaigns worldwide. Autoimmun Rev 2016; 15:1054-1061. [DOI: 10.1016/j.autrev.2016.07.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022]
|
43
|
Herpesvirus Infections and Transglutaminase Type 2 Antibody Positivity in Childhood: The Generation R Study. J Pediatr Gastroenterol Nutr 2016; 63:423-30. [PMID: 26881413 DOI: 10.1097/mpg.0000000000001163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Persistent viral infections have been implicated in the etiology of autoimmune diseases in adulthood, but it is not known whether herpesviruses are associated with the development of celiac disease autoimmunity in childhood. We assessed whether herpesvirus infections are associated with transglutaminase type 2 antibody (TG2A) concentrations in children at 6 years of age. METHODS The present study was embedded within a population-based prospective cohort study. Serum immunoglobulin G levels against Epstein-Barr virus, cytomegalovirus (CMV), and herpes simplex virus type 1 were measured by enzyme-linked immunosorbent assay , and TG2A concentrations with fluorescence enzyme immunoassay in 4420 children at 6 years of age. Children were categorized based on TG2A concentrations into negative (<7 U/mL), positive (≥7-70 U/mL), and strongly positive (≥70 U/mL), that is, 10 times upper limit normal. RESULTS Fifty-nine children (1.3%) were TG2A positive, and of these 31 (53%) had concentrations 70 U/mL or more. Children with TG2A concentrations 70 U/mL or more were less often infected with CMV (adjusted odds ratio (aOR) 0.38, 95% CI 0.14-0.98, P = 0.04) and with any of the 3 viruses (aOR 0.38, 95% CI 0.18-0.78, P < 0.01) than children with TG2A negative concentrations. In addition, children with TG2A concentrations 70 U/mL or more were less often infected with 2 or more viruses than children with TG2A negative concentrations (aOR 0.15, 95% CI 0.03-0.65, P = 0.01). CONCLUSIONS Both CMV single infection and combined CMV, Epstein-Barr virus and/or herpes simplex virus type 1 infections are inversely associated with strongly TG2A positivity. This may indicate a protective effect of herpesvirus infections in the pathogenesis of celiac disease autoimmunity.
Collapse
|
44
|
Agarwal S, Kovilam O, Zach TL, Agrawal DK. Immunopathogenesis and therapeutic approaches in pediatric celiac disease. Expert Rev Clin Immunol 2016; 12:857-69. [PMID: 26999328 PMCID: PMC4975578 DOI: 10.1586/1744666x.2016.1168294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/16/2016] [Indexed: 01/06/2023]
Abstract
Celiac Disease is an autoimmune enteropathy with increasing incidence worldwide in both adults and children. It occurs as an inflammatory condition with destruction of the normal architecture of villi on consumption of gluten and related protein products found in wheat, barley and rye. However, the exact pathogenesis is not yet fully understood. A gluten-free diet remains the main modality of therapy to date. While some patients continue to have symptoms even on a gluten-free diet, adherence to this diet is also difficult, especially for the children. Hence, there is continued interest in novel methods of therapy and the current research focus is on the promising novel non-dietary modalities of treatment. Here, we critically reviewed the existing literature regarding the pathogenesis of celiac disease in children including the role of in-utero exposure leading to neonatal and infant sensitization and its application for the development of new therapeutic approaches for these patients.
Collapse
Affiliation(s)
- Shreya Agarwal
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Oormila Kovilam
- Department of Obstetrics and Gynecology, Creighton University School of Medicine, Omaha, NE, USA
| | - Terence L. Zach
- Department of Pediatrics, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
45
|
Dahan S, Shor DBA, Comaneshter D, Tekes-Manova D, Shovman O, Amital H, Cohen AD. All disease begins in the gut: Celiac disease co-existence with SLE. Autoimmun Rev 2016; 15:848-53. [PMID: 27295421 DOI: 10.1016/j.autrev.2016.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/07/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Case reports and case series have indicated a possible association between celiac disease (CD) and systemic lupus erythematosus (SLE), but additional population-based studies are required. The true prevalence of CD in SLE patients is still unknown, but is indeed an important factor when considering the clinical implications, notably the necessity of screening strategies in SLE patients. Our objective was to investigate the association between CD and SLE using a community-based approach in a real-life population database. METHODS Patients with SLE were compared with age- and sex-matched controls regarding the prevalence of CD in a case-control study. Chi-square and t-tests were used for univariate analysis and a logistic regression model was used for multivariate analysis. The study was performed utilizing the medical database of Clalit Health Services. RESULTS The study included 5018 patients with SLE and 25,090 age- and sex-matched controls. The prevalence of CD was significantly higher in patients with SLE than in controls in univariate analysis (0.8% and 0.2%, respectively, p<0.001). Also, SLE was associated with CD (OR 3.92, 95% CI 2.55-6.03, p<0.001) in a multivariate logistic regression model. CONCLUSIONS Patients with SLE had a greater prevalence of CD than matched controls in a large case-control study. A complex combination of genetic, immunological and novel environmental factors may explain this positive association. Physicians should keep in mind that CD can be a tricky diagnosis in SLE patients, yet a treatable condition, probably more common in this population than we used to think.
Collapse
Affiliation(s)
- Shani Dahan
- Department of Medicine 'B', Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Dana Ben-Ami Shor
- Department of Medicine 'B', Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Israel; Department of Gastroenterology, Sheba Medical Center affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | | | - Dorit Tekes-Manova
- Chaim Sheba General Hospital, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ora Shovman
- Department of Medicine 'B', Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Howard Amital
- Department of Medicine 'B', Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Israel.
| | - Arnon D Cohen
- Chief Physician's Office, Clalit Health Services, Tel Aviv, Israel; Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
46
|
Abstract
OBJECTIVES To assess the prevalence and clinical presentation of celiac disease (CD) in a cohort of children with HLA-DQ2 positive and evaluate the risk factors in the development of CD. METHODS Between July 2004 and July 2005, parents of all healthy full-term newborns in our hospital were invited to participate. HLA-DQ2 was tested in blood sample of the umbilical cord. A point of contact serological test was performed on children between 2 and 3 years of age. Positive results were confirmed by serum anti-transglutaminase 2 and endomysial antibodies. Children with high autoantibody titers underwent an intestinal biopsy. Children of the cohort diagnosed with CD before the screening study were included. Sex, mode of delivery, breast-feeding duration, and age of gluten introduction were studied. RESULTS Of 1291 children, 362 were HLA-DQ2 positive and 262 participated in the study. CD was diagnosed in 4.1% (95% confidence interval (CI) 1.9-6.3). In the whole cohort, 60% had gastrointestinal symptoms, 7% poor weight gain, and 33% were asymptomatic. Five children with potential CD and 6 with CD autoimmunity became negative (42.3%) and are still negative after 5 to 7 years. Female sex was at-risk factor odds ratio 5.7 (95% CI 1.5-20.9), whereas breast-feeding during gluten introduction had a protective effect odds ratio 0.1 (95% CI 0.01-0.8). CONCLUSIONS Prevalence of CD in this cohort was 4%, half of whom had digestive symptoms. Because a high proportion of children showed a spontaneous disappearance of antibodies, prevalence studies of CD in young children should be based on intestinal damage so as not to overestimate results.
Collapse
|
47
|
Nistal E, Caminero A, Herrán AR, Pérez-Andres J, Vivas S, Ruiz de Morales JM, Sáenz de Miera LE, Casqueiro J. Study of duodenal bacterial communities by 16S rRNA gene analysis in adults with active celiac disease vs non-celiac disease controls. J Appl Microbiol 2016; 120:1691-700. [PMID: 26913982 DOI: 10.1111/jam.13111] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 02/03/2016] [Accepted: 02/18/2016] [Indexed: 12/22/2022]
Abstract
AIMS Several studies have suggested that abnormalities in the small-intestinal microbiota might be involved in the development or the pathogenesis of celiac disease (CD). The objective of this study was to characterize and compare the composition of the duodenal microbiota between CD patients and non-CD controls. METHOD AND RESULTS Bacterial communities were identified by pyrosequencing of 16S rRNA extracted from duodenal biopsies. The sequences analysis showed that the majority of the reads were classified within two phyla: Firmicutes and Proteobacteria. Bacterial richness and diversity were higher in non-CD controls than in untreated CD patients, but the differences were not statistically significant. The principal coordinates analysis revealed that bacterial communities of non-CD controls and untreated CD patients were dispersed without forming a clear group according to diagnosis of CD. CONCLUSIONS There are no statistically significant differences in the upper small intestinal composition of bacterial communities between untreated CD patients and non-CD controls. SIGNIFICANCE AND IMPACT OF THE STUDY This pyrosequencing analysis reveals a global picture of the duodenal microbiota that could be useful in future trials investigating the role of the microbiota in CD.
Collapse
Affiliation(s)
- E Nistal
- Área de Microbiología, Facultad de Biología y Ciencias Ambientales, Universidad de León, León, Spain
| | - A Caminero
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, León, Spain
| | - A R Herrán
- Área de Microbiología, Facultad de Biología y Ciencias Ambientales, Universidad de León, León, Spain
| | - J Pérez-Andres
- Área de Microbiología, Facultad de Biología y Ciencias Ambientales, Universidad de León, León, Spain
| | - S Vivas
- Departamento de Gastroenterología, Hospital de León, León, Spain.,Instituto de Biomedicina (IBIOMED), Universidad de León, León, Spain
| | - J M Ruiz de Morales
- Instituto de Biomedicina (IBIOMED), Universidad de León, León, Spain.,Departamento de Inmunología, Hospital de León, León, Spain
| | - L E Sáenz de Miera
- Área de Genética, Facultad de Biología y Ciencias Ambientales, Universidad de León, León, Spain
| | - J Casqueiro
- Área de Microbiología, Facultad de Biología y Ciencias Ambientales, Universidad de León, León, Spain.,Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, León, Spain
| |
Collapse
|
48
|
Temajo NO, Howard N. The virus-induced HSPs regulate the apoptosis of operatus APCs that result in autoimmunity, not in homeostasis. Immunol Res 2015; 60:208-18. [PMID: 25403694 DOI: 10.1007/s12026-014-8585-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The viruses stand salient as environmental factors that trigger autoimmunity. The virus realizes its effects through induction of heat-shock proteins (HSPs) as well as by the viral IE-axis-mediated conversion of organ epithelial cells into virgin de novo professional antigen-presenting cells (APCs). The HSP is the accomplished operator in homeostasis by the logic of it being the regulator of apoptosis. By virtue of its regulation of apoptosis, the HSP is also involved in autoimmunity: (1) adornment of viral IE-axis-generated virgin de novo professional APCs with HSP-induced co-stimulatory molecules which transform these otherwise epithelial cells to competent antigen presenters, the operatus APCs, liable to apoptosis that becomes the initiator of organ damages; (2) molecular mimicry mechanism: epitopes on the HSP may be mistaken for viral peptides and be presented by operatus APCs to autoreactive TCRs resulting in the apoptosis of the operatus APCs; (3) regulation of MHC class II DR-mediated apoptosis of operatus APCS which can result in organ-specific autoimmune syndromes. We should remember, however, that Nature's intended purpose for apoptosis of the professional APCs is benevolence: as a principal regulator of immune homeostasis. But the apoptosis of our postulated operatus APCs can result in autoimmunity. The transformation of virgin de novo professional APCs to operatus APCs mirrors the maturation of DCs through their acquisition of HSP-induced costimulatory molecules. What happens to mature DCs as antigen presenters that end in homeostasis is replicated by what happens to operatus APCs that ends instead in autoimmunity.
Collapse
Affiliation(s)
- Norbert O Temajo
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia,
| | | |
Collapse
|
49
|
Pagliari D, Urgesi R, Frosali S, Riccioni ME, Newton EE, Landolfi R, Pandolfi F, Cianci R. The Interaction among Microbiota, Immunity, and Genetic and Dietary Factors Is the Condicio Sine Qua Non Celiac Disease Can Develop. J Immunol Res 2015; 2015:123653. [PMID: 26090475 PMCID: PMC4451297 DOI: 10.1155/2015/123653] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/14/2014] [Indexed: 12/24/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy, triggered by dietary wheat gluten and similar proteins of barley and rye in genetically susceptible individuals. This is a complex disorder involving both environmental and immune-genetic factors. The major genetic risk factor for CD is determined by HLA-DQ genes. Dysfunction of the innate and adaptive immune systems can conceivably cause impairment of mucosal barrier function and development of localized or systemic inflammatory and autoimmune processes. Exposure to gluten is the main environmental trigger responsible for the signs and symptoms of the disease, but exposure to gluten does not fully explain the manifestation of CD. Thus, both genetic determination and environmental exposure to gluten are necessary for the full manifestation of CD; neither of them is sufficient alone. Epidemiological and clinical data suggest that other environmental factors, including infections, alterations in the intestinal microbiota composition, and early feeding practices, might also play a role in disease development. Thus, this interaction is the condicio sine qua non celiac disease can develop. The breakdown of the interaction among microbiota, innate immunity, and genetic and dietary factors leads to disruption of homeostasis and inflammation; and tissue damage occurs. Focusing attention on this interaction and its breakdown may allow a better understanding of the CD pathogenesis and lead to novel translational avenues for preventing and treating this widespread disease.
Collapse
Affiliation(s)
- D. Pagliari
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - R. Urgesi
- Gastroenterology and Digestive Endoscopy Unit, Belcolle Hospital, 01100 Viterbo, Italy
| | - S. Frosali
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - M. E. Riccioni
- Digestive Endoscopy Unit, Catholic University, 00168 Rome, Italy
| | | | - R. Landolfi
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - F. Pandolfi
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - R. Cianci
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| |
Collapse
|
50
|
Sourdough fermentation of wheat flour does not prevent the interaction of transglutaminase 2 with α2-gliadin or gluten. Nutrients 2015; 7:2134-44. [PMID: 25816160 PMCID: PMC4425136 DOI: 10.3390/nu7042134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 03/19/2015] [Indexed: 01/28/2023] Open
Abstract
The enzyme transglutaminase 2 (TG2) plays a crucial role in the initiation of celiac disease by catalyzing the deamidation of gluten peptides. In susceptible individuals, the deamidated peptides initiate an immune response leading to celiac disease. Several studies have addressed lactic fermentation plus addition of enzymes as a means to degrade gluten in order to prevent adverse response in celiacs. Processing for complete gluten degradation is often harsh and is not likely to yield products that are of comparable characteristics as their gluten-containing counterparts. We are concerned that incomplete degradation of gluten may have adverse effects because it leads to more available TG2-binding sites on gluten peptides. Therefore, we have investigated how lactic acid fermentation affects the potential binding of TG2 to gluten protein in wheat flour by means of estimating TG2-mediated transamidation in addition to measuring the available TG2-binding motif QLP, in α2-gliadin. We show that lactic fermentation of wheat flour, as slurry or as part of sourdough bread, did not decrease the TG2-mediated transamidation, in the presence of a primary amine, to an efficient level (73%–102% of unfermented flour). Nor did the lactic fermentation decrease the available TG2 binding motif QLP in α2-gliadin to a sufficient extent in sourdough bread (73%–122% of unfermented control) to be useful for celiac safe food.
Collapse
|