1
|
Di Tola M, Bontkes HJ, Irure-Ventura J, López-Hoyos M, Bizzaro N. The follow-up of patients with celiac disease. J Transl Autoimmun 2025; 10:100278. [PMID: 39981115 PMCID: PMC11840481 DOI: 10.1016/j.jtauto.2025.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025] Open
Abstract
Celiac disease (CD) is a very common immune-mediated enteropathy resulting from the interaction between dietary gluten and the immune system in genetically predisposed individuals. The immune response leads to intestinal damage, malabsorption and, ultimately, to a broad spectrum of both intestinal and extra-intestinal symptoms. According to current criteria, a proper diagnosis of CD requires an initial phase consisting of clinical case identification and serological screening that, over time, has increased in importance. In most adults and in selected children, the diagnosis is subsequently defined by histological evidence of intestinal damage as a confirmatory test, which usually returns to normal after a suitable period of a gluten-free diet (GFD). The clinical remission and disappearance of circulating antibodies after a GFD further confirm the diagnosis and represent a goal to be achieved to improve the quality of life and reduce the risk of long-term complications. However, although the diagnostic criteria for CD are well defined and described in specific guidelines, the monitoring of CD patients undergoing GFD has been less studied and, consequently, specific guidelines for this phase are still lacking. The aim of this report was to evaluate the classical tools used to monitor the adherence and response to GFD, other non-invasive biomarkers that have been proposed for CD monitoring, and the histological follow-up of CD patients in order to provide a starting point for future discussions on this specific topic.
Collapse
Affiliation(s)
- Marco Di Tola
- UOC Clinical Pathology, San Giovanni - Addolorata Hospital, Rome, Italy
| | - Hetty J. Bontkes
- Department of Laboratory Medicine, Laboratory Specialized Diagnostics and Research, Section Medical Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Juan Irure-Ventura
- Immunology Department, University Hospital Marqués de Valdecilla, Santander, Spain
- Immunopathology Group, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Department, University Hospital Marqués de Valdecilla, Santander, Spain
- Immunopathology Group, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, Azienda Sanitaria Universitaria Integrata, Udine, Italy
| |
Collapse
|
2
|
Yilmaz F, Atay K. A new histomorphological finding in the follow-up of celiac disease: Intraepithelial lymphocyte localization is a reliable indicator of dietary compliance. Ann Diagn Pathol 2025; 75:152438. [PMID: 39813753 DOI: 10.1016/j.anndiagpath.2025.152438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
The correlation between clinical, serological, and endoscopic findings and histological response after a gluten-free diet (GFD) is limited in adult celiac (CD) patients. This study aims to evaluate the effects of GFD on intraepithelial lymphocyte (IEL) localization by comparing the histopathological, clinical, serological, and endoscopic findings of adult CD patients. The patients (n = 131) were divided into three groups: those with good (CDgc) (n = 23) and poor (CDpc) (n = 21) GFD compliance and newly diagnosed ones (nCD) (n = 87). Total and supranuclear IELs were counted per 100 enterocytes and divided into three groups: apical, mixed, and basal, according to ROC (Receiver operating characteristic) analysis. The roles of clinicopathological parameters in predicting good dietary compliance were calculated using the multivariable logistic regression model. CDgc group predominantly (78.3 %) exhibited a basal pattern, and none exhibited an apical. Conversely, most CDpc and nCD patients showed mixed (66.7 % and 73.6 %, respectively) and apical (9.5 % and 25.3 %) patterns. Non-atrophic Marsh types (p = 0.040) and basal pattern (p = 0.043) were independent parameters predicting good dietary compliance. This study first showed that IEL localizations can indicate GFD compliance in samples from CD patients. Localization-based examination of IELs can be an additional histological indicator in monitoring GFD compliance and signs of recovery, especially in adult CD patients.
Collapse
Affiliation(s)
- F Yilmaz
- Mardin Training and Research Hospital, Pathology Laboratory, Mardin, Turkey.
| | - K Atay
- Mardin Training and Research Hospital, Adult Gastroenterology Clinic, Mardin, Turkey
| |
Collapse
|
3
|
Gómez-Aguililla S, Farrais S, López-Palacios N, Arau B, Senosiain C, Corzo M, Fernandez-Jimenez N, Ruiz-Carnicer Á, Fernández-Bañares F, González-García BP, Tristán E, Montero-Calle A, Garranzo-Asensio M, Casado I, Pujals M, Hernández JM, Infante-Menéndez J, Roy G, Sousa C, Núñez C. Diagnosis of celiac disease on a gluten-free diet: a multicenter prospective quasi-experimental clinical study. BMC Med 2025; 23:182. [PMID: 40140887 PMCID: PMC11948932 DOI: 10.1186/s12916-025-04008-y] [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: 10/25/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Diagnosing celiac disease (CD) in individuals adhering to a gluten-free diet (GFD) presents significant challenges. Current guidelines recommend a gluten challenge (GC) lasting at least 6-8 weeks, which has several limitations. Our aim was to compare four approaches previously proposed for diagnosing CD on a GFD: IL-2 serum levels, gut-homing CD8+ T cells, % TCRγδ+ intraepithelial lymphocytes (IELs), and UBE2L3 gene expression. Additionally, we evaluated the CD8+ T-cell-based method with a 3-day GC against the standard GC protocol. METHODS We conducted a multicenter prospective quasi-experimental clinical study. Two subsets of individuals were considered: (1) 20 patients with CD previously diagnosed and 15 non-CD controls, to evaluate the first aim; (2) 41 individuals with uncertain diagnosis who were on a GFD and required GC following current clinical guidelines, to assess the second aim. All participants underwent a 3-day GC (10 g gluten/day). RESULTS Among CD patients and non-CD controls, the sensitivity and specificity of IL-2, gut-homing CD8+ T cells, and UBE2L3 were 82.4% and 83.3%, 88.2% and 100%, and 52.9% and 100%, respectively. The percentage of TCRγδ+ IELs showed 88.2% sensitivity. In the uncertain diagnosis group, a CD8+ T-cell positive response was observed in 8 of the 41 subjects. CONCLUSIONS The percentage of TCRγδ+ IELs and the analysis of IL-2 levels and gut-homing CD8+ T cells are promising diagnostic methods for CD on a GFD. Notably, our results suggest that the CD8+ T-cell assay may provide a consistent and reliable alternative to the extended GC, eliminating the need for invasive procedures to obtain duodenal samples and prolonged gluten ingestion. However, further research with larger cohorts are necessary to validate these findings and establish their definitive clinical utility.
Collapse
Affiliation(s)
- Sara Gómez-Aguililla
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain.
| | - Sergio Farrais
- Servicio de Aparato Digestivo, Hospital Universitario Fundación Jiménez Díaz, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, 28040, Spain
| | - Natalia López-Palacios
- Servicio de Aparato Digestivo, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Beatriz Arau
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Carla Senosiain
- Servicio de Aparato Digestivo, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - María Corzo
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Nora Fernandez-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute, University of the Basque Country (UPV/EHU), Leioa, Basque Country, Spain
| | - Ángela Ruiz-Carnicer
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, 41012, Spain
| | - Fernando Fernández-Bañares
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Bárbara P González-García
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute, University of the Basque Country (UPV/EHU), Leioa, Basque Country, Spain
| | - Eva Tristán
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Ana Montero-Calle
- Functional Proteomics Unit, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - María Garranzo-Asensio
- Functional Proteomics Unit, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Isabel Casado
- Servicio de Anatomía Patológica, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Mar Pujals
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Juana María Hernández
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Jorge Infante-Menéndez
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Garbiñe Roy
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | - Carolina Sousa
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, 41012, Spain
| | - Concepción Núñez
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain.
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Madrid, 28029, Spain.
| |
Collapse
|
4
|
Dodero VI, Herrera MG. Oligomerization of 33-mer Gliadin Peptides: Supramolecular Assemblies in Celiac Disease. ChemMedChem 2025; 20:e202400789. [PMID: 39635969 DOI: 10.1002/cmdc.202400789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/07/2024]
Abstract
The 33-mer gliadin peptide and its deamidated derivative, known as 33-mer DGP, are proteolytically resistant peptides central to the pathomechanism of celiac disease (CeD), the autoimmune presentation of gluten-related disorders (GRD). Both peptides can form spontaneous oligomers in the nanomolar concentration, leading to the formation of nanostructures. In other protein-related diseases, oligomers and aggregates are central in their pathomechanism; therefore, it was hypothesized that the oligomerization of proteolytical-resistant 33-mer gliadin peptides could be an underrecognized disease trigger. This review focuses on the current understanding of 33-mer peptides and their oligomers in vitro and cellular experiments. We intend to give the necessary details that incentivize the chemistry community to get involved in the effort to understand the self-assembly of gliadin peptides and the role of their supramolecular structures in CeD and the other GRD. More research is needed to design effective and safe chemical and/or nutritional interventions beyond the gluten-free diet.
Collapse
Affiliation(s)
- Verónica I Dodero
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - María G Herrera
- Molecular Cell Biology, Faculty of Medicine, Ruhr University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| |
Collapse
|
5
|
Iversen R, Heggelund JE, Das S, Høydahl LS, Sollid LM. Enzyme-activating B-cell receptors boost antigen presentation to pathogenic T cells in gluten-sensitive autoimmunity. Nat Commun 2025; 16:2387. [PMID: 40064932 PMCID: PMC11894174 DOI: 10.1038/s41467-025-57564-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Autoantibodies against the enzyme transglutaminase 3 (TG3) are characteristic to the gluten-sensitive skin disorder dermatitis herpetiformis (DH), which is an extraintestinal manifestation of celiac disease. We here demonstrate that TG3-specific B cells can activate gluten-specific CD4+ T cells through B-cell receptor (BCR)-mediated internalization of TG3-gluten enzyme-substrate complexes. Stereotypic anti-TG3 antibodies using IGHV2-5/IGKV4-1 gene segments enhance the catalytic activity of TG3, and this effect translates into increased gluten presentation to T cells when such antibodies are expressed as BCRs. The crystal structure of TG3 bound to an IGHV2-5/IGKV4-1 Fab shows that antibody binding to a β-sheet in the catalytic core domain causes the enzyme to adopt the active conformation. This mechanism explains the production of stereotypic anti-TG3 autoantibodies in DH and highlights a role for TG3-specific B cells as antigen-presenting cells for gluten-specific T cells. Similar boosting effects of autoreactive BCRs could be relevant for other autoimmune diseases, including rheumatoid arthritis.
Collapse
Affiliation(s)
- Rasmus Iversen
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Julie Elisabeth Heggelund
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Saykat Das
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Lene S Høydahl
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Nextera AS, Oslo, Norway
| | - 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
|
6
|
Dobani S, Kirsty Pourshahidi L, Ternan NG, McDougall GJ, Pereira-Caro G, Bresciani L, Mena P, Almutairi TM, Crozier A, Tuohy KM, Del Rio D, Gill CIR. A review on the effects of flavan-3-ols, their metabolites, and their dietary sources on gut barrier integrity. Food Funct 2025; 16:815-830. [PMID: 39807528 DOI: 10.1039/d4fo04721d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Impairment of gut barrier integrity is associated with the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease, colorectal cancer, and coeliac disease. While many aspects of diet have been linked to improved barrier function, (poly)phenols, a broad group of bioactive phytochemicals, are of potential interest. The (poly)phenolic sub-class, flavan-3-ols, have been investigated in some detail owing to their abundance in commonly consumed foods, including grapes, tea, apples, cocoa, berries, and nuts. This review summarises studies on the effects of flavan-3-ols, their microbiome-mediated metabolites, and food sources of these compounds, on gut barrier structure. Extensive evidence demonstrates that flavan-3-ol rich foods, individual flavan-3-ols (e.g., (epi)catechin, epi(gallo)catechin-3-O-gallate, and pro(antho)cyanidins), and their related microbiota-mediated metabolites, could be effective in protecting and restoring the integrity of the gut barrier. In this context, various endpoints are assessed, including transepithelial electrical resistance of the epithelial layer and expression of tight junction proteins and mucins, in ex vivo, in vitro, and animal models. The differences in bioactivity reported for barrier integrity are structure-function dependent, related to the (poly)phenolic source or the tested compound, as well as their dose, exposure time, and presence or absence of a stressor in the experimental system. Overall, these results suggest that flavan-3-ols and related compounds could help to maintain, protect, and restore gut barrier integrity, indicating that they might contribute to the beneficial properties associated with the intake of their dietary sources. However, rigorous and robustly designed human intervention studies are needed to confirm these experimental observations.
Collapse
Affiliation(s)
- Sara Dobani
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - L Kirsty Pourshahidi
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Nigel G Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Gordon J McDougall
- Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee, UK
| | - Gema Pereira-Caro
- Department of Agroindustry and Food Quality, IFAPA-Alameda Del Obispo, Córdoba, Spain
| | - Letizia Bresciani
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Alan Crozier
- Department of Chemistry, King Saud University Riyadh, Saudi Arabia
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Kieran M Tuohy
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | - Chris I R Gill
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| |
Collapse
|
7
|
Lee AR, Dennis M, Lebovits J, Welstead L, Verma R, Therrien A, Lebwohl B. Dietary assessments in individuals living with coeliac disease: key considerations. J Hum Nutr Diet 2025; 38:e13380. [PMID: 39501424 PMCID: PMC11589401 DOI: 10.1111/jhn.13380] [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: 04/02/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Coeliac disease (CeD) is a type of enteropathy characterised by an immune-mediated reaction to ingested gluten, resulting in impaired absorption of nutrients and symptoms such as bloating, abdominal cramping and diarrhoea. Currently, the only treatment for CeD is adherence to a gluten-free diet (GFD). The latest draft guidance from the US Food and Drug Administration recommends that dietitians experienced in CeD management evaluate patients during the screening and treatment period of CeD clinical trials to assess adherence to a GFD. However, there are currently no standardised guidelines on dietary assessment of patients with CeD on a GFD and there is a lack of widespread availability of expertise in this field. METHODS Based on the findings of a literature review conducted between April and September 2023, this article provides an overview of key points to consider in the nutritional and dietary assessment of patients with CeD who are following a GFD, with particular focus on the clinical trial setting. RESULTS Based on a consensus from dietitians and gastroenterologists experienced in treating patients with CeD, we present specific recommendations for registered dietitians who manage patients with CeD. We also describe the development of a simplified tool for assessment of adherence to a GFD, the Gluten-Free Adherence Survey, based on these recommendations. CONCLUSIONS These guidelines cover nutritional and dietary assessment of patients with CeD, physical assessments, intake of oats, environmental considerations and the disease burden.
Collapse
Affiliation(s)
- Anne R. Lee
- Department of Medicine, Celiac Disease Center, Columbia University Medical CenterColumbia UniversityNew YorkNew YorkUSA
| | - Melinda Dennis
- Division of GastroenterologyCeliac Center, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Jessica Lebovits
- Department of Medicine, Celiac Disease Center, Columbia University Medical CenterColumbia UniversityNew YorkNew YorkUSA
| | - Lori Welstead
- Divisions of Pediatric and Adult Gastroenterology, Hepatology, and Nutrition, Celiac Disease CenterUniversity of ChicagoChicagoIllinoisUSA
| | - Ritu Verma
- Divisions of Pediatric and Adult Gastroenterology, Hepatology, and Nutrition, Celiac Disease CenterUniversity of ChicagoChicagoIllinoisUSA
| | - Amelie Therrien
- Division of GastroenterologyCeliac Center, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Benjamin Lebwohl
- Department of Medicine, Celiac Disease Center, Columbia University Medical CenterColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
8
|
Repo M, Koskimaa S, Paavola S, Kurppa K. Serological testing for celiac disease in children. Expert Rev Gastroenterol Hepatol 2025; 19:155-164. [PMID: 39893645 DOI: 10.1080/17474124.2025.2462245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Celiac disease is one of the most common chronic immune-mediated gastrointestinal conditions, characterized by the presence of disease-specific serum antibodies against self-antigen transglutaminase 2. Even though modern serological assays can identify most untreated celiac disease patients and are also increasingly being used to establish a diagnosis, several challenges are associated with the tests, including a lack of standardization, the variable sensitivity and specificity of commercial assays, and inadequate sensitivity for monitoring adherence to a gluten-free diet. AREAS COVERED This narrative review outlines the current use of serological tests in case-finding and screening, as well as in the follow-up of dietary treatment. Additionally, the possible challenges and pitfalls of serological tests, along with future directions, are addressed. EXPERT OPINION The excellent accuracy of modern autoantibody tests, especially for greatly elevated levels of transglutaminase 2 antibodies and positive endomysial antibodies, enables using serological testing in establishing a diagnosis. However, better international standardization of the assays is required, the necessity of endomysial antibody testing needs to be further scrutinized, and additional research is needed to improve noninvasive tools for follow-up and to further expand the no-biopsy criteria for celiac disease.
Collapse
Affiliation(s)
- Marleena Repo
- Department of Pediatrics, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Sara Koskimaa
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Saana Paavola
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Kalle Kurppa
- Department of Pediatrics, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
- The University Consortium of Seinäjoki, Seinäjoki, Finland
| |
Collapse
|
9
|
Shiha MG, Oka P, Nandi N, Ingham K, Raju SA, Penny HA, Elli L, Sanders DS, Sidhu R. Role of capsule endoscopy and double-balloon enteroscopy in the management of adult patients with coeliac disease and persisting symptoms. Dig Liver Dis 2025; 57:206-212. [PMID: 39095248 DOI: 10.1016/j.dld.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Small bowel capsule endoscopy (CE) and double-balloon enteroscopy (DBE) are recommended for the management of patients with nonresponsive or refractory coeliac disease (CD). However, there is a paucity of data regarding the clinical profiles and outcomes of patients undergoing these investigations. METHODS We conducted a retrospective analysis of two databases of adult patients with CD who underwent CE and/or DBE between 2017 and 2022 at the National Centre for Refractory CD in England. Patient demographic, clinical and endoscopic data were collected, and clinically relevant outcomes were reported. RESULTS A total of 132 patients (median age 53 years, 64.4 % female) underwent 146 CEs and 25 DBEs. The most common symptoms were diarrhoea (51.5 %), abdominal pain (37.8 %), bloating (34.8 %), and weight loss (29.5 %). The overall detection rate of CE and DBE was 87.6 % and 92 %, respectively. Following CE and DBE, 14 patients (10.6 %) were diagnosed with CD-related complications such as ulcerative jejunitis, strictures and malignancy. Seven patients (5.3 %) died during follow-up, with five of these deaths directly attributed to CD. Older age, weight loss and anaemia were associated with poor outcomes. CONCLUSIONS The sequential approach of CE and DBE identified CD-related complications in almost 1 in 10 patients with nonresponsive or refractory CD. Older patients with persistent villous atrophy, weight loss and anaemia require close monitoring to help with the early diagnosis and management of complications.
Collapse
Affiliation(s)
- Mohamed G Shiha
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK.
| | - Priya Oka
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Nicoletta Nandi
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy
| | - Katerina Ingham
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Suneil A Raju
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Hugo A Penny
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - David S Sanders
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Reena Sidhu
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
10
|
Chen WH, Hsu CC, Ho HJ, Smith J, Smith S, Huang HY, Chang HC, Hsiao YC. Rapid Gluten Allergen Detection Using an Integrated Photoimaging Assay and Ionic Liquid Extraction Sensor. ACS OMEGA 2024; 9:49767-49777. [PMID: 39713688 PMCID: PMC11656241 DOI: 10.1021/acsomega.4c08411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024]
Abstract
In recent years, food allergies and food sensitivities have remained critical public health problems that affect approximately 15% of the global population. Wheat is a major food source worldwide, but it is also a common food allergen. Celiac disease is chronic immune-mediated enteropathy triggered by exposure to dietary gluten in genetically predisposed individuals; it can be treated only through strict gluten avoidance. Therefore, rapid gluten detection is crucial for protecting the health of patients. Gluten contains two primary water-insoluble proteins: gliadin and glutenin. Gliadin is a key contributor to celiac disease and poses challenges for sample pretreatment owing to its insolubility, thereby reducing the accuracy and sensitivity of detection systems. Rapid sample processing is a critical problem in gliadin detection. In this report, we developed a gliadin sensor system called the integrated food allergy and microorganism sensor (iFAMs). The iFAMs comprises a gliadin lateral flow chip, a one-pot extraction solution, and an image assay app. The iFAMs enables gliadin extraction and detection in under 2 min with high sensitivity (0.04 mg/kg for gliadin, lower than the regulatory limit of 20 mg/kg). Users can easily measure gluten concentrations in samples and quantify gliadin levels using the smartphone-based image assay app. In samples collected from restaurants, the iFAMs successfully detected hidden gluten within "gluten-free" food items. The compact size and user-friendly design of the iFAMs render it suitable for not only consumers but also clinicians, food industries, and regulators to enhance food safety.
Collapse
Affiliation(s)
- Wen-Hao Chen
- Research
and Development Group, Leo Verification
Systems Inc., Powell, Wyoming 82435, United States
- School
of
Biological Sciences, Nanyang Technological
University, 639798 Singapore
| | - Chuan-Chih Hsu
- Department
of Surgery, College of Medicine, Taipei
Medical University, Taipei 110, Taiwan
- Department
of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Hsin-Jung Ho
- Technology
Commercialization Center, Taipei Medical
University, Taipei 110, Taiwan
| | - Jill Smith
- Research
and Development Group, Leo Verification
Systems Inc., Powell, Wyoming 82435, United States
| | - Seaton Smith
- Research
and Development Group, Leo Verification
Systems Inc., Powell, Wyoming 82435, United States
| | - Hui-Yin Huang
- Research
and Development Group, Leo Verification
Systems Inc., Powell, Wyoming 82435, United States
| | | | - Yu-Cheng Hsiao
- Graduate
Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan
- Cell Physiology
and Molecular Image Research Center, Taipei
Medical University, Taipei 110, Taiwan
- School
of
Biological Sciences, Nanyang Technological
University, 639798 Singapore
| |
Collapse
|
11
|
Kori M, Gabbai A, Shamir R, Guz-Mark A. Children with celiac disease, diagnosed with or without biopsy, present similar adherence to gluten-free diet and serology decline. Eur J Pediatr 2024; 184:21. [PMID: 39549103 DOI: 10.1007/s00431-024-05849-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/18/2024]
Abstract
Current professional guidelines enable diagnosing pediatric Celiac Disease (CeD) without a biopsy, when tissue transglutaminase (TTG) IgA antibodies are > × 10 the upper limit of normal (ULN) and anti-endomysial antibodies (EMA) are positive in a second sample. We compared baseline characteristics and serology normalization in children diagnosed with or without biopsies. A retrospective study of pediatric patients diagnosed with CeD during 2020: group A, no biopsy and group B, biopsy-based diagnosis. Baseline characteristics included demographics, anthropometrics, symptoms, family history, and celiac serology. Follow-up at 6-month intervals, up to 18 months, included dietary compliance, symptoms, and serology. Of 145 children diagnosed with CeD, 42 (29%) and 103 (71%) were from group A and B respectively. Mean age was 7.8 years (range 2.4-17.9 y), 91 (62.8%) females. Baseline symptoms or signs were present in 93 (64.1%) children, without significant difference between the groups. Baseline TTG levels were > × 10 ULN in all patients in group A and 71 (68.9%) in group B. Among these patients, the rate of TTG decline during follow-up did not differ at any time point between patients diagnosed with and without biopsy, and between patients with and without symptoms. At the last follow-up visit, 24 (57%) children in group A and 46 (65%) in group B had TTG < × 3 ULN without significant difference between the groups. CONCLUSION Rate of TTG decline did not differ between CeD patients diagnosed with and without biopsy, suggesting that, at least in short term, no biopsy approach may not change patients' adherence and families' attitude towards treatment. WHAT IS KNOWN • Based on current guidelines, there is a rise in the incidence of pediatric celiac disease (CeD) diagnosis without an intestinal biopsy. • There is insufficient data regarding patients' adherence to treatment, including pattern of serology decline, based on the method of CeD diagnosis. WHAT IS NEW • Children with CeD have similar baseline characteristics, including presence or absence of symptoms, whether diagnosed with or without biopsies. • During 18-month follow-up, the rate of celiac serology decline, and the reported adherence to treatment, do not differ between patients diagnosed based on biopsy or no biopsy approaches.
Collapse
Affiliation(s)
- Michal Kori
- Pediatric Gastroenterology, Kaplan Medical Center, Rehovot, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Assaf Gabbai
- Division of Pediatrics, Kaplan Medical Center, Rehovot, Israel
| | - Raanan Shamir
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Anat Guz-Mark
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
12
|
Stanciu D, Staykov H, Dragomanova S, Tancheva L, Pop RS, Ielciu I, Crișan G. Gluten Unraveled: Latest Insights on Terminology, Diagnosis, Pathophysiology, Dietary Strategies, and Intestinal Microbiota Modulations-A Decade in Review. Nutrients 2024; 16:3636. [PMID: 39519469 PMCID: PMC11547711 DOI: 10.3390/nu16213636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A decade of research on gluten-related disorders (GRDs) is reviewed in this study, with a particular emphasis on celiac disease (CD) and non-celiac gluten sensitivity (NCGS). GRDs are triggered by the ingestion of gluten and gluten-like proteins found in wheat, barley, and rye. These proteins lead to intestinal damage in celiac disease, an autoimmune condition characterized by villous atrophy and a variety of gastrointestinal and extraintestinal symptoms. More enigmatic and less understood, NCGS involves symptoms similar to CD but without the immunological reaction or intestinal damage. Recent years have seen advances in the understanding of GRDs, particularly in connection to how intestinal microbiota influences disease progression and patient outcomes. The gluten-free diet (GFD) is still the standard therapy recommended for GRDs despite significant challenges, as discussed in this article. Precise diagnostic methods, patient education and dietary counseling are critical for improving patients' quality of life. The purpose of this review is to provide a more clear and up-to-date understanding of GRDs, and to help further research on this important topic.
Collapse
Affiliation(s)
- Dana Stanciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Hristian Staykov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Radu Samuel Pop
- 3rd Department of Pediatrics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania;
| | - Irina Ielciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Gianina Crișan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| |
Collapse
|
13
|
Dayon L, Núñez Galindo A, Chevalier J, Aquarius M, Otten B, Troost FJ, Duncan P, Affolter M. Detection of gluten peptides in human duodenal fluids with immuno-liquid chromatography-mass spectrometry. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6819-6828. [PMID: 39264106 DOI: 10.1039/d4ay00852a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Gluten proteins are storage proteins in wheat that exhibit a certain resistance to gastrointestinal digestion. To explore solutions to cope with accidental ingestion of gluten in individuals suffering from gluten-related disorders, it is essential to monitor the fate of gluten peptides in biological samples, i.e., gastrointestinal juices, blood plasma or urine. In this work, we aimed at developing a mass spectrometry (MS)-based method for measuring gluten peptides in human duodenal fluids. Seven gluten peptides, including the well-documented 33-mer gluten peptide (LQLQPFPQPQLPYPQPQLPYPQPQLPYPQPQPF), were selected after a literature review and characterization of a gluten-containing product. Isotopically labelled peptides were used as references and a targeted liquid chromatography (LC) MS assay based on high resolution parallel reaction monitoring (PRM) was designed. Despite iterative and fine tuning of the LC-PRM-MS method, the low level of endogenous gluten peptides in human duodenal fluid samples precluded their direct detection. Thus, an initial immunoprecipitation (IP) step was included. Several antibodies were tested, and one proved reliable for the enrichment of the 33-mer gluten peptide as well as a few additional gluten peptides. Figures-of-merits of the immuno-LC-PRM-MS assay were assessed with a focus on quantification trueness and precision. We have developed an MS-based method for measuring the 33-mer gluten peptide in human duodenal fluids. Based on isotopic dilution, the method relies on the combination of IP and LC-PRM-MS analysis. Measurements were shown to be sensitive, quantitative, and reproducible.
Collapse
Affiliation(s)
- Loïc Dayon
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
| | - Julien Chevalier
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Michèl Aquarius
- Department of Gastroenterology, VieCuri Medical Center, Venlo, The Netherlands
| | - Britt Otten
- Department of Human Biology, NUTRIM, Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation (HEFI), Maastricht University, Venlo, The Netherlands
| | - Freddy J Troost
- Department of Human Biology, NUTRIM, Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation (HEFI), Maastricht University, Venlo, The Netherlands
| | - Peter Duncan
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Michael Affolter
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
14
|
Vereczkei Z, Szakács Z, Peresztegi MZ, Lemes K, Hagymási K, Dakó S, Dakó E, Lada S, Faluhelyi N, Szekeres G, Pásztor G, Farkas N, Pár G, Mezősi E, Bajor J. Influence of a structured, 1-year-long dietary intervention regarding body composition and cardiovascular risk (ARCTIC) in coeliac disease: a protocol of a multicentre randomised controlled trial. BMJ Open 2024; 14:e084365. [PMID: 39384235 PMCID: PMC11474741 DOI: 10.1136/bmjopen-2024-084365] [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: 01/16/2024] [Accepted: 08/30/2024] [Indexed: 10/11/2024] Open
Abstract
INTRODUCTION Coeliac disease (CD) affects 1% of the population worldwide. The only available evidence-based treatment is a strict gluten-free diet (GFD), which can readily lead to weight gain and unfavourable metabolic changes (eg, dyslipidaemia, fatty liver disease and insulin resistance) if followed without adequate dietary control. That can lead to increased cardiovascular risk (CV). We planned a randomised controlled trial to test the effect of a group-based, structured, 1-year, advanced dietary education, per the proposal of a Mediterranean diet vs standard of care, regarding the most relevant CV risk factors (eg, metabolic parameters and body composition) in CD patients. METHODS AND ANALYSIS Randomisation will occur after the baseline dietary education and interview in a 1:1 allocation ratio. Outcomes include anthropometric parameters (body composition analysis including weight, Body Mass Index, fat mass, per cent body fat, skeletal muscle mass, visceral fat area and total body water) and CV risk-related metabolic parameters (eg, lipid profile, homocysteine, fasting glucose, haemoglobin A1c, Homeostatic Model Assessment Index, metabolic hormones, waist circumference, blood pressure, liver function tests, liver steatosis rate and diet composition). In this study, we aim to draw attention to a new aspect regarding managing CD: dietary education can lead to a better quality of the GFD, thereby reducing the risk of potential metabolic and CV complications. ETHICS AND DISSEMINATION The study was approved by the Scientific and Research Ethics Committee of the Hungarian Medical Research Council (27521-5/2022/EÜIG). Findings will be disseminated at research conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05530070.
Collapse
Affiliation(s)
- Zsófia Vereczkei
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsolt Szakács
- First Department of Medicine, Medical Shcool, University of Pécs, Pécs, Hungary
| | | | - Klára Lemes
- Department of Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Krisztina Hagymási
- Department of Surgery, Transplantation and Gastroenterology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Sarolta Dakó
- Department of Surgery, Transplantation and Gastroenterology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Eszter Dakó
- Department of Surgery, Transplantation and Gastroenterology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Szilvia Lada
- Department of Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Nándor Faluhelyi
- Department of Radiology, Medical School, University of Pécs, Pécs, Hungary
| | - Gábor Szekeres
- Department of Radiology, Medical School, University of Pécs, Pécs, Hungary
| | - Gyula Pásztor
- Department of Radiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Nelli Farkas
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Gabriella Pár
- First Department of Medicine, Medical Shcool, University of Pécs, Pécs, Hungary
| | - Emese Mezősi
- First Department of Medicine, Medical Shcool, University of Pécs, Pécs, Hungary
| | - Judit Bajor
- First Department of Medicine, Medical Shcool, University of Pécs, Pécs, Hungary
| |
Collapse
|
15
|
Ge HJ, Chen XL. Advances in understanding and managing celiac disease: Pathophysiology and treatment strategies. World J Gastroenterol 2024; 30:3932-3941. [PMID: 39351055 PMCID: PMC11438662 DOI: 10.3748/wjg.v30.i35.3932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
In this editorial, we comment on an article published in the recent issue of the World Journal of Gastroenterology. Celiac disease (CeD) is a disease occurring in genetically susceptible individuals, which is mainly characterized by gluten intolerance in the small intestine and clinical symptoms such as abdominal pain, diarrhea, and malnutrition. Therefore, patients often need a lifelong gluten-free diet, which greatly affects the quality of life and expenses of patients. The gold standard for diagnosis is intestinal mucosal biopsy, combined with serological and genetic tests. At present, the lack of safe, effective, and satisfactory drugs for CeD is mainly due to the complexity of its pathogenesis, and it is difficult to find a perfect target to solve the multi-level needs of patients. In this editorial, we mainly review the pathological mechanism of CeD and describe the current experimental and improved drugs for various pathological aspects.
Collapse
Affiliation(s)
- Hao-Jie Ge
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
16
|
Rodríguez-Ramírez R, Fernández Peralbo MA, Mendía I, Long JCD, Sousa C, Cebolla Á. Urinary excretion of gluten immunoreactive peptides as an indicator of gastrointestinal function after fasting and dietary provocation in healthy volunteers. Front Immunol 2024; 15:1433304. [PMID: 39161759 PMCID: PMC11330814 DOI: 10.3389/fimmu.2024.1433304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Introduction Understanding intestinal permeability is paramount for elucidating gastrointestinal health and pathology. The size and nature of the molecule traversing the intestinal barrier offer crucial insights into various acute and chronic diseases, as well as the evolution of some conditions. This study aims to assess the urinary excretion kinetics of gluten immunogenic peptides (u-GIP), a unique class of dietary peptides detectable in urine, in volunteers under controlled dietary conditions. This evaluation should be compared to established probes like lactulose, a non-digestible disaccharide indicative of paracellular permeability, and mannitol, reflecting transcellular permeability. Methods Fifteen participants underwent simultaneous ingestion of standardized doses of gluten (10 g), lactulose (10 g), and mannitol (1 g) under fasting conditions for at least 8 hours pre-ingestion and during 6 hours post-ingestion period. Urine samples were collected over specified time intervals. Excretion patterns were analyzed, and correlations between the lactulose-to-mannitol ratio (LMR) and u-GIP parameters were assessed. Results The majority of u-GIP were detected within the first 12 hours post-ingestion. Analysis of the variability in cumulative excretion across two sample collection ranges demonstrated that lactulose and u-GIP exhibited similar onset and excretion dynamics, although GIP reached its maximum peak earlier than either lactulose or mannitol. Additionally, a moderate correlation was observed between the LMR and u-GIP parameters within the longest urine collection interval, indicating potential shared characteristics among permeability pathways. These findings suggest that extending urine collection beyond 6 hours may enhance data reliability. Discussion This study sheds light on the temporal dynamics of u-GIP in comparison to lactulose and mannitol, established probes for assessing intestinal permeability. The resemblance between u-GIP and lactulose excretion patterns aligns with the anticipated paracellular permeability pathway. The capacity to detect antigenic food protein fragments in urine opens novel avenues for studying protein metabolism and monitoring pathologies related to the digestive and intestinal systems.
Collapse
Affiliation(s)
- Raquel Rodríguez-Ramírez
- Research and Development Department, Biomedal S.L., Seville, Spain
- Inorganic Chemistry Department, Faculty of Science, University of Granada, Granada, Spain
| | | | - Irati Mendía
- Research and Development Department, Biomedal S.L., Seville, Spain
| | | | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Ángel Cebolla
- Research and Development Department, Biomedal S.L., Seville, Spain
| |
Collapse
|
17
|
Moreno MDL, González-Rovira M, Martínez-Pancorbo C, Martín-Cameán M, Nájar-Moyano AM, Romero M, de la Hoz E, López-Beltrán C, Mellado E, Bartha JL, Brodin P, Rodríguez-Herrera A, Sainz-Bueno JA, Sousa C. Foetal gluten immunogenic peptides during pregnancy: a new determinant on the coeliac exposome. BMC Med 2024; 22:295. [PMID: 39020299 PMCID: PMC11256569 DOI: 10.1186/s12916-024-03495-9] [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: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The increasing incidence of coeliac disease is leading to a growing interest in active search for associated factors, even the intrauterine and early life. The exposome approach to disease encompasses a life course perspective from conception onwards has recently been highlighted. Knowledge of early exposure to gluten immunogenic peptides (GIP) in utero could challenge the chronology of early prenatal tolerance or inflammation, rather than after the infant's solid diet after birth. METHODS We developed an accurate and specific immunoassay to detect GIP in amniotic fluid (AF) and studied their accumulates, excretion dynamics and foetal exposure resulting from AF swallowing. One hundred twenty-five pregnant women with different gluten diets and gestational ages were recruited. RESULTS GIP were detectable in AF from at least the 16th gestational week in gluten-consuming women. Although no significant differences in GIP levels were observed during gestation, amniotic GIP late pregnancy was not altered by maternal fasting, suggesting closed-loop entailing foetal swallowing of GIP-containing AF and subsequent excretion via the foetal kidneys. CONCLUSIONS The study shows evidence, for the first time, of the foetal exposure to gluten immunogenic peptides and establishes a positive correlation with maternal gluten intake. The results obtained point to a novel physiological concept as they describe a plausible closed-loop circuit entailing foetal swallowing of GIP contained in AF and its subsequent excretion through the foetal kidneys. The study adds important new information to understanding the coeliac exposome.
Collapse
Affiliation(s)
- María de Lourdes Moreno
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - María González-Rovira
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | | | - María Martín-Cameán
- Obstetric and Gynecology Department, High Risk Pregnancy Unit, Autoimmune Diseases and Pregnancy Clinic, University Hospital La Paz, Madrid, Spain
| | - Ana María Nájar-Moyano
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mercedes Romero
- Obstetric and Gynecology Department, Sagrado Corazón Hospital, Seville, Spain
| | - Esther de la Hoz
- Obstetric and Gynecology Department, Sagrado Corazón Hospital, Seville, Spain
| | | | - Encarnación Mellado
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - José Luis Bartha
- Obstetric and Gynecology Department, High Risk Pregnancy Unit, Autoimmune Diseases and Pregnancy Clinic, University Hospital La Paz, Madrid, Spain
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Uppsala, Sweden
| | - Alfonso Rodríguez-Herrera
- St. Luke's General Hospital, UCD School of Medicine, University College Dublin, Kilkenny &, Kilkenny, Ireland
| | - José Antonio Sainz-Bueno
- Department of Obstetrics and Gynecology, Valme University Hospital, Seville, Spain
- Department of Obstetrics and Gynecology, University of Seville, Seville, Spain
| | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
18
|
Martín-Cardona A, Carrasco A, Arau B, Vidal J, Tristán E, Ferrer C, Gonzalez-Puglia G, Pallarès N, Tebé C, Farrais S, Núñez C, Fernández-Bañares F, Esteve M. γδ+ T-Cells Is a Useful Biomarker for the Differential Diagnosis between Celiac Disease and Non-Celiac Gluten Sensitivity in Patients under Gluten Free Diet. Nutrients 2024; 16:2294. [PMID: 39064736 PMCID: PMC11279444 DOI: 10.3390/nu16142294] [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: 06/17/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND The differential diagnosis between patients with celiac disease (CD) and non-celiac gluten sensitivity (NCGS) is difficult when a gluten-free diet (GFD) has been initiated before the diagnostic work-up. Isolated increases in TCRγδ+ and celiac lymphogram (increased TCRγδ+ plus decreased CD3-) may enable differential diagnosis in this challenging clinical setting. This study evaluated: (1) the accuracy of %TCRγδ+ and celiac lymphogram for diagnosing CD before and after GFD and for differentiation with NCGS; (2) TCRγδ+ kinetics at baseline and after starting GFD in both CD and NCGS. METHODS The inclusion criteria were patients with CD (n = 104), NCGS (n = 37), and healthy volunteers (n = 18). An intestinal biopsy for intraepithelial lymphogram by flow cytometry was performed at baseline and after GFD. The optimal cutoff for CD diagnostic accuracy was established by maximizing the Youden index and via logistic regression. RESULTS %TCRγδ+ showed better diagnostic accuracy than celiac lymphogram for identifying CD before and after GFD initiation. With a cutoff > 13.31, the accuracy for diagnosing CD in patients under GFD was 0.88 [0.80-0.93], whereas the accuracy for diagnosing NCGS (%TCRγδ+ ≤ 13.31) was 0.84 [0.76-0.89]. The percentage of TCRγδ+ cells showed differential kinetics between CD (baseline 22.7% [IQR, 16.4-33.6] vs. after GFD 26.4% [IQR, 17.8-36.8]; p = 0.026) and NCGS (baseline 9.4% [IQR, 4.1-14.6] vs. after GFD 6.4% [IQR, 3.2-11]; p = 0.022). CONCLUSION TCRγδ+ T cell assessment accurately diagnoses CD before and after a GFD. Increased TCRγδ+ was maintained in the long term after GFD in CD but not in NCGS. Altogether, this suggests the potential usefulness of this marker for the differential diagnosis of these two entities in patients on a GFD.
Collapse
Affiliation(s)
- Albert Martín-Cardona
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Anna Carrasco
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Beatriz Arau
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Judith Vidal
- Department of Flow Cytometry, Catlab, 08232 Viladecavalls, Spain;
| | - Eva Tristán
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carme Ferrer
- Pathology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain;
| | - Gerardo Gonzalez-Puglia
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
| | - Natàlia Pallarès
- Biostatistics Support and Research Unit, Germans Trias i Pujol Research Institute and Hospital (IGTP), 08916 Badalona, Spain; (N.P.); (C.T.)
| | - Cristian Tebé
- Biostatistics Support and Research Unit, Germans Trias i Pujol Research Institute and Hospital (IGTP), 08916 Badalona, Spain; (N.P.); (C.T.)
| | - Sergio Farrais
- Gastroenterology Department, Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain;
| | - Concepción Núñez
- Laboratorio de Investigación en Genética de Enfermedades Complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Fernando Fernández-Bañares
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Esteve
- Gastroenterology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, 08221 Terrassa, Spain; (A.M.-C.); (A.C.); (B.A.); (E.T.); (G.G.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Ruiz-Carnicer Á, Segura V, Moreno MDL, Coronel-Rodríguez C, Sousa C, Comino I. Transfer of celiac disease-associated immunogenic gluten peptides in breast milk: variability in kinetics of secretion. Front Immunol 2024; 15:1405344. [PMID: 39034995 PMCID: PMC11257844 DOI: 10.3389/fimmu.2024.1405344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background Exposure to antigens is crucial for child immune system development, aiding disease prevention and promoting infant health. Some common food antigen proteins are found in human breast milk. However, it is unclear whether gluten antigens linked to celiac disease (CD) are transmitted through breast milk, potentially impacting the development of the infant's immune system. Objective This study aimed to analyze the passage of gluten immunogenic peptides (GIP) into human breast milk. We evaluated the dynamics of GIP secretion after lactating mothers adopted a controlled gluten-rich diet. Methods We prospectively enrolled 96 non-CD and 23 CD lactating mothers, assessing total proteins and casein in breast milk, and GIP levels in breast milk and urine. Subsequently, a longitudinal study was conducted in a subgroup of 12 non-CD lactating mothers who adopted a controlled gluten-rich diet. GIP levels in breast milk and urine samples were assayed by multiple sample collections over 96 hours. Results Analysis of a single sample revealed that 24% of non-CD lactating mothers on a regular unrestricted diet tested positive for GIP in breast milk, and 90% tested positive in urine, with significantly lower concentrations in breast milk. Nevertheless, on a controlled gluten-rich diet and the collection of multiple samples, GIP were detected in 75% and 100% of non-CD participants in breast milk and urine, respectively. The transfer dynamics in breast milk samples were long-enduring and GIP secretion persisted from 0 to 72 h. In contrast, GIP secretion in urine samples was limited to the first 24 h, with inter-individual variations. In the cohort of CD mothers, 82.6% and 87% tested negative for GIP in breast milk and urine, respectively. Conclusions This study definitively established the presence of GIP in breast milk, with substantial inter-individual variations in secretion dynamics. Our findings provide insights into distinct GIP kinetics observed in sequentially collected breast milk and urine samples, suggesting differential gluten metabolism patterns depending on the organ or system involved. Future research is essential to understand whether GIP functions as sensitizing or tolerogenic agents in the immune system of breastfed infants.
Collapse
Affiliation(s)
- Ángela Ruiz-Carnicer
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Verónica Segura
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - María de Lourdes Moreno
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | | | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Isabel Comino
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| |
Collapse
|
20
|
Lee CC, Suttikhana I, Ashaolu TJ. Techno-Functions and Safety Concerns of Plant-Based Peptides in Food Matrices. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12398-12414. [PMID: 38797944 DOI: 10.1021/acs.jafc.4c02464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Plant-based peptides (PBPs) benefit functional food development and environmental sustainability. Proteolysis remains the primary method of peptide production because it is a mild and nontoxic technique. However, potential safety concerns still emanate from toxic or allergenic sequences, amino acid racemization, iso-peptide bond formation, Maillard reaction, dose usage, and frequency. The main aim of this review is to investigate the techno-functions of PBPs in food matrices, as well as their safety concerns. The distinctive characteristics of PBPs exhibit their techno-functions for improving food quality and functionality by contributing to several crucial food formulations and processing. The techno-functions of PBPs include solubility, hydrophobicity, bitterness, foaming, oil-binding, and water-holding capacities, which subsequently affect food matrices. The safety and quality of foodstuff containing PBPs depend on the proper source of plant proteins, the selection of processing approaches, and compliance with legal regulations for allergen labeling and safety evaluations. The safety concerns in allergenicity and toxicity were discussed. The conclusion is that food technologists must apply safe limits and consider potential allergenic components generated during the development of food products with PBPs. Therefore, functional food products containing PBPs can be a promising strategy to provide consumers with wholesome health benefits.
Collapse
Affiliation(s)
- Chi-Ching Lee
- Department of Food Engineering, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Halkalı Avenue No: 28, Halkalı, Küçükçekmece, Istanbul 34303, Türkiye
| | - Itthanan Suttikhana
- Department of Multifunctional Agriculture, Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, České Budějovice, Branišovská 1645/31a, 370 05 České Budějovice 2, Czechia
| | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang 550000, Viet Nam
- Faculty of Medicine, Duy Tan University, Da Nang 550000, Viet Nam
| |
Collapse
|
21
|
Mello CRLE, da Silva GAP, Lins MTC, Antunes MMDC. Gluten-free diet adherence in pediatric celiac patients: Evaluating CDAT questionnaire and test for gluten detection in urine. J Pediatr Gastroenterol Nutr 2024; 78:1310-1316. [PMID: 38477385 DOI: 10.1002/jpn3.12180] [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: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVES This study aimed to evaluate the contributions of the Adapted Celiac Dietary Adherence Test (CDAT) and the Rapid Urinary Gluten Detection Test (u-GIP) in assessing gluten-free diet adherence in children and adolescents with celiac disease. METHODS Fifty-four celiac patients from two pediatric gastroenterology outpatient clinics affiliated with university hospitals were evaluated. The original CDAT was adapted for children through a transcultural process, and the original cutoff point was adopted to define adherence. A single examiner carried out the u-GIP test in fresh urine samples. Sociodemographic and clinical factors and family food security status were also evaluated. RESULTS A total of 88.9% of participants (confidence interval [CI]: 77.4-95.8; p<0.001) adhered to the gluten-free diet, as determined by the adapted CDAT score, while 87.0% (CI: 75.1-94.6; p<0.001) had negative u-GIP results. Among the 48 children adhering to the CDAT, six exhibited positive u-GIP results in a urine sample. Of the six nonadherent participants, only one had a positive u-GIP result. Notably, none of the children and adolescents with celiac disease who tested positive for u-GIP reported symptoms on the day of testing, and their growth rates remained stable. CONCLUSIONS Even celiac children and adolescents adhering to the CDAT questionnaire may show a positive u-GIP in a single measurement without accompanying symptoms or growth impairment. The u-GIP could be helpful in complementary tests in specific situations, such as for patients who exhibit compliant behavior but still experience symptoms or maintain persistent positive serology.
Collapse
Affiliation(s)
- Carolina R Lins E Mello
- Department of Medical Sciences, Children and Adolescent Health Graduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Gisélia A P da Silva
- Department of Medical Sciences, Children and Adolescent Health Graduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Manuela T C Lins
- Ambulatory of Pediatric Gastroenterology, Instituto de Medicina Integral Professor Fernando Figueira, Recife, Pernambuco, Brazil
| | - Margarida M de C Antunes
- Department of Medical Sciences, Children and Adolescent Health Graduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
22
|
Anderson RP, Verma R, Schumann M. A Look Into the Future: Are We Ready for an Approved Therapy in Celiac Disease? Gastroenterology 2024; 167:183-193. [PMID: 38355059 DOI: 10.1053/j.gastro.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
As it appears that we are currently at the cusp of an era in which drugs that are new, re-purposed, or "supplements" will be introduced to the management of celiac disease, we need to reflect on whether the framework is set for celiac disease to be treated increasingly with pharmaceuticals as well as diet. This refers to reflecting on the rigor of current diagnostic practices; the limitations of the current standard of care, which is a gluten-free diet; and that we lack objective markers of disease severity. Investigating these issues will help us to identify gaps in technology and practices that could be critical for selecting patients with a well-defined need for an improved or alternative treatment. Both aspects, circumscribed limitations of the gluten-free diet and diagnostics helping to define celiac disease target groups, together with the guiding requirements by the responsible regulatory authorities, will contribute to defining the subgroups of patients with confirmed celiac disease eligible for distinct pharmacologic strategies. Because many patients with celiac disease are diagnosed in childhood, these aspects need to be differentially discussed for the pediatric setting. In this perspective, we aimed to describe these contextual issues and then looked ahead to the future. What might be the major challenges in celiac disease clinics in the coming years once drugs are an option alongside diet? And what will be the future objectives for researchers who further decipher the mucosal immunology of celiac disease? Speculating on the answers to these questions is as stimulating as it is fascinating to be part of this turning point.
Collapse
Affiliation(s)
- Robert P Anderson
- Gastroenterology Service, Mackay Base Hospital, West Mackay, Queensland, Australia
| | - Ritu Verma
- University of Chicago, Comer Children's Hospital, Chicago, Illinois
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
23
|
Silvester JA, Elli L, Khosla C, Tye-Din JA. Past, Present, and Future of Noninvasive Tests to Assess Gluten Exposure, Celiac Disease Activity, and End-Organ Damage. Gastroenterology 2024; 167:159-171. [PMID: 38670279 PMCID: PMC11235091 DOI: 10.1053/j.gastro.2024.01.053] [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: 11/17/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 04/28/2024]
Abstract
Although many biomarkers have been proposed, and several are in widespread clinical use, there is no single readout or combination of readouts that correlates tightly with gluten exposure, disease activity, or end-organ damage in treated patients with celiac disease. Challenges to developing and evaluating better biomarkers include significant interindividual variability-related to immune amplification of gluten exposure and how effects of immune activation are manifest. Furthermore, the current "gold standard" for assessment of end-organ damage, small intestinal biopsy, is itself highly imperfect, such that a marker that is a better reflection of the "ground truth" may indeed appear to perform poorly. The goal of this review was to analyze past and present efforts to establish robust noninvasive tools for monitoring treated patients with celiac disease and to highlight emerging tools that may prove to be useful in clinical practice.
Collapse
Affiliation(s)
- Jocelyn A Silvester
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Harvard Celiac Disease Research Program, Harvard Medical School, Boston, Massachusetts.
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chaitan Khosla
- Sarafan ChEM-H, Departments of Chemistry and Chemical Engineering, Stanford University, Stanford, California
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Gastroenterology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Malamut G, Soderquist CR, Bhagat G, Cerf-Bensussan N. Advances in Nonresponsive and Refractory Celiac Disease. Gastroenterology 2024; 167:132-147. [PMID: 38556189 DOI: 10.1053/j.gastro.2024.02.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/04/2024] [Accepted: 02/20/2024] [Indexed: 04/02/2024]
Abstract
Nonresponsive celiac disease (CeD) is relatively common. It is generally attributed to persistent gluten exposure and resolves after correction of diet errors. However, other complications of CeD and disorders clinically mimicking CeD need to be excluded. Novel therapies are being evaluated to facilitate mucosal recovery, which might benefit patients with nonresponsive CeD. Refractory CeD (RCeD) is rare and is divided into 2 types. The etiology of type I RCeD is unclear. A switch to gluten-independent autoimmunity is suspected in some patients. In contrast, type II RCeD represents a low-grade intraepithelial lymphoma. Type I RCeD remains a diagnosis of exclusion, requiring ruling out gluten intake and other nonmalignant causes of villous atrophy. Diagnosis of type II RCeD relies on the demonstration of a clonal population of neoplastic intraepithelial lymphocytes with an atypical immunophenotype. Type I RCeD and type II RCeD generally respond to open-capsule budesonide, but the latter has a dismal prognosis due to severe malnutrition and frequent progression to enteropathy-associated T-cell lymphoma; more efficient therapy is needed.
Collapse
Affiliation(s)
- Georgia Malamut
- Department of Gastroenterology, Assistance Publique-Hôpitaux de Paris Centre-Université Paris Cité, Hôpital Cochin, Paris, France; Laboratory of Intestinal Immunity, INSERM UMR 1163-Institut Imagine, Université Paris Cité, Paris, France.
| | - Craig R Soderquist
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, INSERM UMR 1163-Institut Imagine, Université Paris Cité, Paris, France.
| |
Collapse
|
25
|
Haimi M, Lerner A. Utilizing Telemedicine Applications in Celiac Disease and Other Gluten-Free-Diet-Dependent Conditions: Insights from the COVID-19 Pandemic. Healthcare (Basel) 2024; 12:1132. [PMID: 38891207 PMCID: PMC11171739 DOI: 10.3390/healthcare12111132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Globally, approximately 1.4% of people have celiac disease (CD), induced by gluten sensitivity. If left untreated, it causes small intestinal inflammation and villous atrophy, which can result in failure to thrive, anemia, osteoporosis, malabsorption, and even malignancy. The only treatment option available is a gluten-free diet (GFD). Few studies have looked at the role and perception of telehealth in relation to CD and selective nutrition both before and after the COVID-19 pandemic. AIM Our goal was to screen and investigate the research conducted both before and after the COVID-19 pandemic concerning the utilization of telehealth applications and solutions in CD and other GFD-dependent circumstances. METHODS We employed a narrative review approach to explore articles that were published in scholarly journals or organizations between the years 2000 and 2024. Only English-language publications were included. PubMed and Google Scholar searches were mainly conducted using the following keywords: telemedicine, telehealth, telecare, eHealth, m-health, COVID-19, SARS-CoV-2, celiac disease, and gluten-free diet (GFD). Manual searches of the references in the acquired literature were also carried out, along with the authors' own personal contributions of their knowledge and proficiency in this field. RESULTS Only a few studies conducted prior to the COVID-19 outbreak examined the viewpoints and experiences of adult patients with CD with relation to in-person clinic visits, as well as other options such as telehealth. The majority of patients believed that phone consultations were appropriate and beneficial. Video conferencing and telemedicine became more popular during the COVID-19 pandemic, demonstrating the effectiveness of using these technologies for CD on a global basis. In recent years, urine assays for gluten identification have become accessible for use at home. These tests could be helpful for CD monitoring with telemedicine assistance. CONCLUSIONS The extended knowledge gathered from the COVID-19 pandemic is expected to complement pre-COVID-19 data supporting the usefulness of telemedicine even after the emergent pandemic, encouraging its wider adoption in standard clinical practice. The monitoring and follow-up of CD patients and other GFD-dependent conditions can greatly benefit from telemedicine.
Collapse
Affiliation(s)
- Motti Haimi
- Health Systems Management Department, The Max Stern Yezreel Valley College, Yezreel Valley 1930600, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Ramat Gan 5266202, Israel;
- Research Department, Ariel University, Ariel 407000, Israel
| |
Collapse
|
26
|
Herrera MG, Amundarain MJ, Dörfler PW, Dodero VI. The Celiac-Disease Superantigen Oligomerizes and Increases Permeability in an Enterocyte Cell Model. Angew Chem Int Ed Engl 2024; 63:e202317552. [PMID: 38497459 DOI: 10.1002/anie.202317552] [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: 11/17/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024]
Abstract
Celiac disease (CeD) is an autoimmune disorder triggered by gluten proteins, affecting approximately 1 % of the global population. The 33-mer deamidated gliadin peptide (DGP) is a metabolically modified wheat-gluten superantigen for CeD. Here, we demonstrate that the 33-mer DGP spontaneously assembles into oligomers with a diameter of approximately 24 nm. The 33-mer DGP oligomers present two main secondary structural motifs-a major polyproline II helix and a minor β-sheet structure. Importantly, in the presence of 33-mer DGP oligomers, there is a statistically significant increase in the permeability in the gut epithelial cell model Caco-2, accompanied by the redistribution of zonula occludens-1, a master tight junction protein. These findings provide novel molecular and supramolecular insights into the impact of 33-mer DGP in CeD and highlight the relevance of gliadin peptide oligomerization.
Collapse
Affiliation(s)
- Maria G Herrera
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, C1428EG, Argentina
| | - Maria J Amundarain
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - Philipp W Dörfler
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - Veronica I Dodero
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| |
Collapse
|
27
|
Perez-Junkera G, Ruiz de Azua L, Vázquez-Polo M, Lasa A, Fernandez Gil MP, Txurruka I, Navarro V, Larretxi I. Global Approach to Follow-Up of Celiac Disease. Foods 2024; 13:1449. [PMID: 38790748 PMCID: PMC11119929 DOI: 10.3390/foods13101449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Celiac disease, an autoimmune disorder induced by the ingestion of gluten, affects approximately 1.4% of the population. Gluten damages the villi of the small intestine, producing symptoms such as abdominal pain, bloating and a subsequent loss of nutrient absorption, causing destabilization of the nutritional status. Moreover, gluten can trigger extra intestinal symptoms, such as asthma or dermatitis, but also mental disorders such as depression or anxiety. Moreover, people suffering from celiac disease sometimes feel misunderstood by society, mainly due to the lack of knowledge about the disease and the gluten-free diet. Thus, the treatment and follow-up of patients with celiac disease should be approached from different perspectives, such as the following: (1) a clinical perspective: symptomatology and dietary adherence monitorization; (2) nutritional assessment: dietary balance achievement; (3) psychological assistance: mental disorders avoidance; and (4) social inclusion: educating society about celiac disease in order to avoid isolation of those with celiac disease. The aim of this narrative review is to gain deep insight into the different strategies that currently exist in order to work on each of these perspectives and to clarify how the complete approach of celiac disease follow-up should be undertaken so that the optimum quality of life of this collective is reached.
Collapse
Affiliation(s)
- Gesala Perez-Junkera
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Children’s National Hospital 111 Michigan Avenue NW, Washington, DC 20010, USA
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
| | - Lorea Ruiz de Azua
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
| | - Maialen Vázquez-Polo
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
| | - Arrate Lasa
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
| | - María Pilar Fernandez Gil
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
| | - Itziar Txurruka
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
| | - Virginia Navarro
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
| | - Idoia Larretxi
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain; (G.P.-J.); (L.R.d.A.); (M.V.-P.); (M.P.F.G.); (I.T.); (V.N.); (I.L.)
- Bioaraba, Nutrición y Seguridad Alimentaria, 01006 Vitoria-Gasteiz, Spain
- Centro Integral de Atención a Mayores San Prudencio, Ayuntamiento de Vitoria-Gasteiz, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
28
|
Raju SA, Shiha MG, Penny HA. Monitoring coeliac disease in 2024, time to change practice? Curr Opin Gastroenterol 2024; 40:190-195. [PMID: 38547329 DOI: 10.1097/mog.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Persistent villous atrophy is associated with morbidity in coeliac disease and most commonly due to ongoing gluten ingestion. Current methods for assessing gluten exposure and persisting villous atrophy include dietary questionnaires and repeat duodenal biopsy, which have limited accuracy or are invasive. This review discusses adjunctive and/or novel tests that could be used to overcome these challenges. RECENT FINDINGS Small bowel capsule endoscopy is well tolerated and helps to evaluate for persisting villous atrophy and importantly, complications associated with coeliac disease. Testing for urinary and/or stool gluten immunogenic peptides may help identify recent gluten exposure, but further studies are still warranted to evaluate the accuracy and applicability of this approach. Measuring spikes in circulating Interleukin-2 following gluten challenge has shown promise for coeliac disease diagnosis, and thus may serve as a useful confirmatory test in those with persisting symptoms but provides no information on mucosal inflammation. No specific gut microbial signature has been identified in coeliac disease; however, studies have shown a reduced microbial diversity in active disease, which with future refinement may prove clinically useful. SUMMARY There is no evidence to support alternative methods for assessing persisting villous atrophy in coeliac disease over performing an up-to-date duodenal biopsy. Monitoring for adherence to a gluten-free diet remains clinically challenging and should be a priority for future research.
Collapse
Affiliation(s)
- Suneil A Raju
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Mohamed G Shiha
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Hugo A Penny
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
29
|
Abstract
Celiac disease (CeD) is a chronic immune-mediated enteropathy, which occurs in genetically predisposed individuals by the ingestion of gluten proteins present in wheat, barley and rye. The global pooled prevalence of CeD is 0.7% and it has been reported from nations all around the globe and can affect individuals of any age. It has a wide clinical spectrum ranging from being asymptomatic to being symptomatic with severe manifestations. Though initial descriptions of CeD focused on the classical presentation with gastrointestinal manifestations, in recent years it has been found that more patients have non-classical manifestations such as anemia, osteoporosis, increased transaminases, failure to thrive or short stature. The definitive diagnosis of CeD is based on a combination of clinical history, serologic testing with/without examination of duodenal biopsies. The preferred initial serologic test regardless of age for the detection of CeD is the tissue transglutaminase (IgA anti-tTG). Children with a high tTG-IgA (≥10 ULN) AND a positive anti-endomysial IgA antibody (EMA) can be diagnosed to have CeD without the need for duodenal biopsies. The rest should undergo biopsies with at least 4 biopsies from the distal duodenum and at least 1 from the bulb. A correctly orientated biopsy showing increased intraepithelial cells and a villous to crypt ratio of <2 is suggestive of CeD. The management of CeD is a lifelong complete dietary avoidance of gluten. IgA-TGA acts as a surrogate marker for healing of the small-bowel mucosa and should be performed every 6 mo until normalization and then every 12-24 mo thereafter.
Collapse
Affiliation(s)
- Rishi Bolia
- Department of Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, 501, Stanley Street, South Brisbane, QLD, 4101, Australia.
| | - Nikhil Thapar
- Department of Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, 501, Stanley Street, South Brisbane, QLD, 4101, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
- Woolworths Centre for Child Nutrition Research, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
30
|
Mazzola AM, Zammarchi I, Valerii MC, Spisni E, Saracino IM, Lanzarotto F, Ricci C. Gluten-Free Diet and Other Celiac Disease Therapies: Current Understanding and Emerging Strategies. Nutrients 2024; 16:1006. [PMID: 38613039 PMCID: PMC11013189 DOI: 10.3390/nu16071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
A lifelong gluten-free diet (GFD) is the only treatment for celiac disease and other gluten-related disorders. Nevertheless, strict adherence to the GFD is often challenging due to concerns about social isolation, risk of gluten contaminations, high cost, poor quality and the taste of gluten-free products. Moreover, although the GFD is effective in achieving mucosal healing, it may lead to dietary imbalances due to nutrient deficiencies over a long period of time. To overcome these issues, several gluten-free wheat flours have been developed to create products that closely resemble their gluten-containing counterparts. Furthermore, given the critical importance of adhering to the GFD, it becomes essential to promote adherence and monitor possible voluntary or involuntary transgressions. Various methods, including clinical assessment, questionnaires, serology for celiac disease, duodenal biopsies and the detection of Gluten Immunogenic Peptides (GIPs) are employed for this purpose, but none are considered entirely satisfactory. Since adherence to the GFD poses challenges, alternative therapies should be implemented in the coming years to improve treatment efficacy and the quality of life of patients with celiac disease. The aim of this narrative review is to explore current knowledge of the GFD and investigate its future perspectives, focusing on technology advancements, follow-up strategies and insights into a rapidly changing future.
Collapse
Affiliation(s)
- Anna Maria Mazzola
- Gastroenterology Unit, Spedali Civili Hospital, 25123 Brescia, Italy; (A.M.M.); (I.Z.); (F.L.)
- Department of Internal Medicine and Medical Therapy, University of Pavia, 27100 Pavia, Italy
| | - Irene Zammarchi
- Gastroenterology Unit, Spedali Civili Hospital, 25123 Brescia, Italy; (A.M.M.); (I.Z.); (F.L.)
| | - Maria Chiara Valerii
- Unit of Translational Physiology and Nutrition, Department of Biological, Geological and Environmental Sciences, University of Bologna, 40126 Bologna, Italy; (M.C.V.); (E.S.)
| | - Enzo Spisni
- Unit of Translational Physiology and Nutrition, Department of Biological, Geological and Environmental Sciences, University of Bologna, 40126 Bologna, Italy; (M.C.V.); (E.S.)
| | - Ilaria Maria Saracino
- Microbiology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Francesco Lanzarotto
- Gastroenterology Unit, Spedali Civili Hospital, 25123 Brescia, Italy; (A.M.M.); (I.Z.); (F.L.)
| | - Chiara Ricci
- Gastroenterology Unit, Spedali Civili Hospital, 25123 Brescia, Italy; (A.M.M.); (I.Z.); (F.L.)
- Department of Experimental and Clinical Science, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
31
|
Meyer N, Illarionov B, Fischer M, Wieser H. Preparation and Immunochemical Characterization of a Water-Soluble Gluten Peptide Fraction for Improving the Diagnosis of Celiac Disease. Nutrients 2024; 16:742. [PMID: 38474870 DOI: 10.3390/nu16050742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The diagnosis of celiac disease (CD) is complex and requires a multi-step procedure (symptoms, serology, duodenal biopsy, effect of a gluten-free diet, and optional genetic). The aim of the study was to contribute to the improvement of CD diagnosis by preparing a water-soluble gluten peptide fraction (called Solgluten) and by selecting gluten-specific enzyme-linked immunosorbent assays (ELISA) for the detection of gluten immunogenic gluten peptides (GIPs) in urine and blood serum spiked with Solgluten. Food-grade Solgluten was prepared by the extraction of a peptic digest of vital gluten with water, centrifugation, and freeze-drying. The process was relatively easy, repeatable, and cheap. The content of gliadin-derived GIPs was 491 mg/g. Solgluten was used as antigenic material to compare two competitive ELISA kits (R7021 and K3012) and two sandwich ELISA kits (M2114 and R7041) in their quality regarding the quantitation of GIPs in urine and blood serum. The quality parameters were the reactivity, sensitivity, coefficients of variation and determination, and curve shape. The evaluation of the kits showed a number of discrepancies in individual quality parameters measured in urine and serum. Due to the lowest limit of quantitation and the highest coefficient of determination, M2114 may be the first choice, while R7021 appeared to be less suitable because of the high coefficients of variation and unfavorable curve progression. The results set the stage for improving CD diagnosis by supplementing conventional blood tests with oral provocation with Solgluten and subsequent ELISA measurement of GIPs that could support the no-biopsy approach and by better assessing the effect of a gluten-free diet by monitoring adherence to the diet by measuring GIPs in urine and blood.
Collapse
Affiliation(s)
- Niklas Meyer
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, 20146 Hamburg, Germany
| | - Boris Illarionov
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, 20146 Hamburg, Germany
| | - Markus Fischer
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, 20146 Hamburg, Germany
| | - Herbert Wieser
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
32
|
Elli L, Leffler D, Cellier C, Lebwohl B, Ciacci C, Schumann M, Lundin KEA, Chetcuti Zammit S, Sidhu R, Roncoroni L, Bai JC, Lee AR, Dennis M, Robert ME, Rostami K, Khater S, Comino I, Cebolla A, Branchi F, Verdu EF, Stefanolo JP, Wolf R, Bergman-Golden S, Trott N, Scudeller L, Zingone F, Scaramella L, Sanders DS. Guidelines for best practices in monitoring established coeliac disease in adult patients. Nat Rev Gastroenterol Hepatol 2024; 21:198-215. [PMID: 38110546 DOI: 10.1038/s41575-023-00872-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 12/20/2023]
Abstract
Coeliac disease (CeD) is an immunological disease triggered by the consumption of gluten contained in food in individuals with a genetic predisposition. Diagnosis is based on the presence of small bowel mucosal atrophy and circulating autoantibodies (anti-type 2 transglutaminase antibodies). After diagnosis, patients follow a strict, life-long gluten-free diet. Although the criteria for diagnosis of this disease are well defined, the monitoring phase has been studied less and there is a lack of specific guidelines for this phase. To develop a set of clinical guidelines for CeD monitoring, we followed the Grading of Recommendations Assessment, Development and Evaluation methodology. Statements and recommendations with the level of evidence were developed and approved by the working group, which comprised gastroenterologists, pathologists, dieticians and biostatisticians. The proposed guidelines, endorsed by the North American and European coeliac disease scientific societies, make recommendations for best practices in monitoring patients with CeD based on the available evidence. The evidence level is low for many topics, suggesting that further research in specific aspects of CeD would be valuable. In conclusion, the present guidelines support clinicians in improving CeD treatment and follow-up and highlight novel issues that should be considered in future studies.
Collapse
Affiliation(s)
- Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease-Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Daniel Leffler
- Celiac Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Christophe Cellier
- Department of Gastroenterology and Endoscopy, CELAC network, AP-HP Centre, Hôpital Européen Georges Pompidou, Université de Paris, Cité and Institut National du Cancer, Paris, France
| | - Benjamin Lebwohl
- Celiac Disease Center, Department of Medicine, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Carolina Ciacci
- Center for Celiac Disease, Gastrointestinal Unit, AOU San Giovanni di Dio e Ruggi D'Aragona and Department of Medicine Surgery Dentistry, Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Michael Schumann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Berlin, Germany
| | - Knut E A Lundin
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | | | - Reena Sidhu
- Department of Infection, Immunity and Cardiovascular Diseases, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease-Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Julio C Bai
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires, Argentina
| | - Anne R Lee
- Celiac Disease Center, Department of Medicine, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Melinda Dennis
- Celiac Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Marie E Robert
- Department of Pathology and Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kamran Rostami
- Department of Gastroenterology, Palmerston North District Health Board (DHB), Palmerston North, New Zealand
| | - Sherine Khater
- Department of Gastroenterology and Endoscopy, CELAC network, AP-HP Centre, Hôpital Européen Georges Pompidou, Université de Paris, Cité and Institut National du Cancer, Paris, France
| | - Isabel Comino
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | | | - Federica Branchi
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Berlin, Germany
| | - Elena F Verdu
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Juan Pablo Stefanolo
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires, Argentina
| | - Randi Wolf
- Program in Nutrition, Department of Health Studies & Applied Educational Psychology, Teachers College, Columbia University, New York, NY, USA
| | - Sheba Bergman-Golden
- Program in Nutrition, Department of Health Studies & Applied Educational Psychology, Teachers College, Columbia University, New York, NY, USA
| | - Nick Trott
- Department of Infection, Immunity and Cardiovascular Diseases, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Luigia Scudeller
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Gastroenterology Unit, Azienda Ospedale-Università Padova, Padua, Italy
| | - Lucia Scaramella
- Center for Prevention and Diagnosis of Celiac Disease-Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - David S Sanders
- Department of Infection, Immunity and Cardiovascular Diseases, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| |
Collapse
|
33
|
Russell AK, Lucas EC, Henneken LM, Pizzey CJ, Clarke D, Myleus A, Tye-Din JA. Stool Gluten Peptide Detection Is Superior to Urinary Analysis, Coeliac Serology, Dietary Adherence Scores and Symptoms in the Detection of Intermittent Gluten Exposure in Coeliac Disease: A Randomised, Placebo-Controlled, Low-Dose Gluten Challenge Study. Nutrients 2024; 16:279. [PMID: 38257173 PMCID: PMC10818827 DOI: 10.3390/nu16020279] [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: 12/14/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Monitoring adherence to a gluten-free diet is an important goal of coeliac disease management. Urine and stool gluten immunogenic peptide (GIP) assays provide an objective readout of gluten ingestion, with the former favoured due to its convenience and acceptability. This study assessed stool GIP excretion after low-dose gluten challenge designed to mimic accidental gluten exposure. A total of 52 coeliac participants undertook a randomised, double-blind gluten (50-1000 mg) or placebo challenge. Stool and urinary GIP, serology, dietary adherence and symptoms were assessed. Stool GIP was 100% sensitive for gluten intake ≥250 mg and 71% for 50 mg. Peak GIP detection was 12-36 h after gluten exposure. The mean stool GIP after 1000 mg gluten ingestion remained above the limit of quantification for 5 days. Urine GIP assessment had poor sensitivity for GIP excretion compared to stool. Serology, dietary adherence score and symptoms did not correlate with gluten excretion during lead-in. We conclude that stool GIP detection is highly sensitive, with levels related to gluten dose and time from ingestion. Weekly or bi-weekly testing will detect low-level exposure more effectively than urine GIP assessments or traditional methods. In this seronegative, apparently well-treated cohort, a high frequency of baseline-positive GIP suggests ongoing gluten exposure, but the assessment of patient behaviour and assay specificity is needed.
Collapse
Affiliation(s)
- Amy K. Russell
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (A.K.R.)
| | - Erin C. Lucas
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (A.K.R.)
| | - Lee M. Henneken
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Catherine J. Pizzey
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Dean Clarke
- National Measurement Institute, Port Melbourne, VIC 3207, Australia
| | - Anna Myleus
- Department of Public Health and Clinical Medicine, Family Medicine, Umea University, 901 87 Umea, Sweden
| | - Jason A. Tye-Din
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (A.K.R.)
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- The Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| |
Collapse
|
34
|
Zingone F, Zanini A. Gluten free diet, assessment of its adherence, and quality of life. PEDIATRIC AND ADULT CELIAC DISEASE 2024:189-198. [DOI: 10.1016/b978-0-443-13359-6.00011-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Jafari E, Soleymani N, Hamidi M, Rahi A, Rezaei A, Azizian R. Celiac Disease: A Review from Genetic to Treatment. IRANIAN BIOMEDICAL JOURNAL 2024; 28:8-14. [PMID: 38444380 PMCID: PMC10994635 DOI: 10.61186/ibj.4028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/29/2023] [Indexed: 03/07/2024]
Abstract
Celiac disease (CD) is a complex disorder influenced by genetic and environmental factors. When people with a genetic predisposition to CD consume gluten, an inflammatory response is triggered in the small intestine, and this reaction can be alleviated by the elimination of gluten from the diet. The clinical manifestations of CD vary greatly from person to person and begin at a young age or in adulthood. Influence of genetic factors on CD development is evident in carriers of the DQ2 and/or DQ8 allele. HLA genotypes are associated with gut colonization by bacteria, particularly in individuals suffering from CD. In addition, beneficial gut microbes are crucial for the production of DPP-4, which plays a key role in immune function, as well as metabolic and intestinal health. Therefore, probiotics have been recommended as a complementary food supplement in CD.
Collapse
Affiliation(s)
- Erfaneh Jafari
- Pediatric Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Soleymani
- Department of Food Hygiene, Islamic Azad University (Science and Research Branch), Tehran, Iran
| | - Masoud Hamidi
- École Polytechnique de Bruxelles-BioMatter Unit, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Azar Rahi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Rezaei
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Azizian
- Pediatric Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Biomedical Innovation and Start-Up Association (Biomino), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Guandalini S, Sansotta N. Celiac disease in pediatric patients. PEDIATRIC AND ADULT CELIAC DISEASE 2024:77-101. [DOI: 10.1016/b978-0-443-13359-6.00010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Volta U, Caio G, De Giorgio R. Serology and screening in celiac disease. PEDIATRIC AND ADULT CELIAC DISEASE 2024:125-137. [DOI: 10.1016/b978-0-443-13359-6.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Crepaldi M, Palo M, Maniero D, Bertin L, Savarino EV, Anderson RP, Zingone F. Emerging Pharmaceutical Therapies to Address the Inadequacy of a Gluten-Free Diet for Celiac Disease. Pharmaceuticals (Basel) 2023; 17:4. [PMID: 38275990 PMCID: PMC10821495 DOI: 10.3390/ph17010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Celiac disease (CeD) is a chronic autoimmune disorder triggered by the ingestion of gluten, affecting around 1% of the global population. It is a multifactorial disease involving both genetics and environmental factors. Nowadays, the only available treatment for CeD is a life-long gluten-free diet (GFD), which can cause a significant burden for patients, since symptoms and mucosal injury can persist despite apparent compliance with a GFD. This could also lead to psychological consequences and affect the quality of life of these patients. Thankfully, recent advances in understanding the pathogenesis of CeD and the availability of various targets have made it feasible to explore pharmaceutical treatments specific to CeD. Recently, the FDA has highlighted the unmet needs of adult patients on a GFD who experience ongoing symptoms attributed to CeD and also show persistent duodenal villous atrophy. This review will outline the limitations of a GFD, describe the targets of potential novel treatment of CeD and provide an overview of the primary clinical trials involving oral and injectable agents for a non-dietary treatment of CeD.
Collapse
Affiliation(s)
- Martina Crepaldi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Michela Palo
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Robert P. Anderson
- Gastroenterology Department, Mackay Base Hospital, Mackay, QLD 4740, Australia
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| |
Collapse
|
39
|
Studerus D, Schumann M. [What is established in the management of celiac disease?]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023; 64:1162-1170. [PMID: 37962594 DOI: 10.1007/s00108-023-01621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
Nowadays, celiac disease is well-established in internal medicine as an autoimmune disease induced by gluten as a trigger. Undoubtedly similarly well-established is the gluten-free diet. It is the only recognized therapy for celiac disease to date. However, this presents some pitfalls in its implementation, which will be discussed in the following review. In addition, current developments that have the potential to significantly change both diagnosis and treatment of celiac disease are discussed. On the one hand, such an outlook was chosen since colleagues want to be "ready" when such developments are integrated into daily clinical routine. On the other hand, the realization that the field of mucosal immunology is moving forward has the potential to lift the spirits of the reader.
Collapse
Affiliation(s)
- Diana Studerus
- Zöliakie Zentrum Schweiz & GastroZentrum Hirslanden, Zürich, Schweiz
| | - Michael Schumann
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Deutschland.
| |
Collapse
|
40
|
Scarmozzino F, Pizzi M, Pelizzaro F, Angerilli V, Dei Tos AP, Piazza F, Savarino EV, Zingone F, Fassan M. Refractory celiac disease and its mimickers: a review on pathogenesis, clinical-pathological features and therapeutic challenges. Front Oncol 2023; 13:1273305. [PMID: 38023263 PMCID: PMC10662059 DOI: 10.3389/fonc.2023.1273305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Refractory celiac disease (RCD) and enteropathy-associated T-cell lymphoma (EATL) are rare, yet severe complications of celiac disease (CD). Over the last decades, several studies have addressed the biology and clinical-pathological features of such conditions, highlighting unique disease patterns and recurrent genetic events. Current classification proposals identify two forms of RCD, namely: (i) type 1 RCD (RCD-I), characterized by phenotypically normal intra-epithelial lymphocytes (IELs); and (ii) type 2 RCD (RCD-II), featuring phenotypically aberrant IELs. While RCD-I likely represents a gluten-independent dysimmune reaction against small bowel epithelial cells, RCD-II is better considered an in situ aggressive T-cell lymphoma, with high rates of progression to overt EATL. The diagnosis of RCD and EATL is often challenging, due to misleading clinical-pathological features and to significant overlap with several CD-unrelated gastro-intestinal disorders. Similarly, the treatment of RCD and EATL is an unmet clinical need for both gastroenterologists and hematologists. Moving from such premises, this review aims to provide a comprehensive view of RCD and EATL, specifically considering their pathogenesis and the many still open issues concerning their diagnosis and clinical management.
Collapse
Affiliation(s)
- Federico Scarmozzino
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Marco Pizzi
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Filippo Pelizzaro
- Gastroenterology Unit, Department of Surgical, Gastroenterological and Oncological Sciences -DISCOG, University of Padua School of Medicine, Padua, Italy
| | - Valentina Angerilli
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Angelo Paolo Dei Tos
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Francesco Piazza
- Hematology & Clinical Immunology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Edoardo Vincenzo Savarino
- Gastroenterology Unit, Department of Surgical, Gastroenterological and Oncological Sciences -DISCOG, University of Padua School of Medicine, Padua, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgical, Gastroenterological and Oncological Sciences -DISCOG, University of Padua School of Medicine, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| |
Collapse
|
41
|
Singh SK, Srivastava A. Transition of Care in Celiac Disease. Indian J Pediatr 2023; 90:1142-1148. [PMID: 37273133 DOI: 10.1007/s12098-023-04611-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/17/2023] [Indexed: 06/06/2023]
Abstract
Celiac disease (CD) is a gluten related disorder which affects all age-groups and occurs in genetically susceptible population after introduction of gluten in diet. The worldwide prevalence of CD is ~1% and it is higher in certain "at-risk groups". The clinical features are variable, ranging from classical diarrhea to an asymptomatic state. Diagnosis requires serology and duodenal histology although a non-biopsy diagnosis is recommended by European Society of Pediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) for a select group of children. Treatment of CD is with a life-long strict gluten free diet (GFD) along with correction of nutritional deficiencies. Regular follow-up to assess compliance and efficacy of GFD is mandatory. Non-responsive CD needs evaluation by a specialist as it can be due to incorrect diagnosis, poor dietary compliance, coexisting conditions like small bowel bacterial overgrowth, pancreatic insufficiency etc. and lastly, refractory CD. Most patients diagnosed as CD in childhood receive no medical or dietary supervision after transition to adulthood and nearly a third are non-compliant to GFD. No requirement of medications, patient's perception of understanding GFD and absence of symptoms with intermittent non-compliance leads to neglect of care after transition. Poor dietary adherence leads to nutritional deficiencies, osteoporosis, fertility issues and risk of malignancy. It is mandatory that the patients know about CD, need of strict GFD, regular follow-up, disease complications, and are capable of communicating with the health-care personnel before transition. Formulating a phased transition care program with joint pediatric and adult clinics is required for a successful transition and improving the long-term outcome.
Collapse
Affiliation(s)
- Sumit K Singh
- Department of Pediatrics, Sri Aurobindo Medical College and Postgraduate Institute, Indore, Madhya Pradesh, India
| | - Anshu Srivastava
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
42
|
Aldzhyan V, Tamamian C, Tabibian JH. Leveraging telemedicine in gastroenterology and hepatology: a narrative review. Mhealth 2023; 9:36. [PMID: 38023778 PMCID: PMC10643195 DOI: 10.21037/mhealth-23-27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Objective Over the years, telemedicine has played a prominent role in delivering healthcare to patients. Due to its flexibility and many benefits, telemedicine confers physicians the ability to guide and promote medical care remotely. The advent of the coronavirus disease 2019 (COVID-19) pandemic has changed the landscape of medicine and has accelerated the usage of digital and remote healthcare systems for clinical care. Herein, we provide an overview of telemedicine, its applications in managing inflammatory bowel disease (IBD), celiac disease (CD), and liver diseases, its advantages and limitations, and its use in educating the next generation of gastroenterologists. Methods We conducted a review of scientific articles published in PubMed and Google Scholar. Articles were selected based on the search terms included in the search strategy summary. The language of the articles was restricted to English only. Key Content and Findings We report that telemedicine has the potential to streamline and improve patient care in gastroenterology (GI) and hepatology while also limiting health care expenses. Additionally, we noted the importance of tele-education for training the next generation of physicians who intend on practicing in rural settings. Furthermore, we identified barriers to telemedicine care that exacerbate health inequities and potential solutions to achieving digital health equity. Lastly, we briefly discuss the role of artificial intelligence (AI) in remote patient monitoring. Conclusions Although telemedicine has existed for many decades, over the past decade there have been many advancements in telemedicine applications in GI and hepatology. Despite its broad benefits, further research needs to be done to alleviate barriers to telemedicine care.
Collapse
Affiliation(s)
- Vahagn Aldzhyan
- Department of Science and Health, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Carine Tamamian
- University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - James H. Tabibian
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Gastroenterology, Adventist Health Glendale Medical Center, Glendale, CA, USA
| |
Collapse
|
43
|
Belei O, Jugănaru I, Basaca DG, Munteanu AI, Mărginean O. The Role of Intestinal Microbiota in Celiac Disease and Further Therapeutic Perspectives. Life (Basel) 2023; 13:2039. [PMID: 37895421 PMCID: PMC10608277 DOI: 10.3390/life13102039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy caused by exposure to gluten and related prolamins in genetically susceptible individuals. It is a complex genetic disorder with multiple contributing genes. Linkage studies have identified several genomic regions that probably contain CD susceptibility genes. The most important genetic factors are HLA-DQ2 and DQ8. Several known environmental triggers promote the onset of CD at any age after gluten introduction in individuals with a genetic background, such as viral infections and intestinal dysbiosis. Recent publications have described the interference of the intestinal microbiome in gluten metabolism, modulation of local immune reactions, and in maintaining normal gut permeability. These results have promoted further lines of research on the benefit of probiotic administration to prevent disease onset or alleviate clinical symptoms along with a gluten-free diet (GFD). The relationship between gut microbiome changes and the onset of CD is incompletely understood, still being the subject of current research. This narrative review analyzes the interplay between environmental factors, intestinal microbiome alterations, and the course of CD. Furthermore, this review sets out to discuss if modulation of intestinal microflora with pre- and probiotics along with a GFD could represent a reliable therapeutic target for celiac patients.
Collapse
Affiliation(s)
- Oana Belei
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Iulius Jugănaru
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Diana-Georgiana Basaca
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Andrei Ioan Munteanu
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Otilia Mărginean
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
44
|
Simón E, Molero-Luis M, Fueyo-Díaz R, Costas-Batlle C, Crespo-Escobar P, Montoro-Huguet MA. The Gluten-Free Diet for Celiac Disease: Critical Insights to Better Understand Clinical Outcomes. Nutrients 2023; 15:4013. [PMID: 37764795 PMCID: PMC10537989 DOI: 10.3390/nu15184013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The gluten-free diet (GFD) remains a complex paradigm in managing celiac disease (CeD) in children and adults, and there are many reasons why GFD adherence should be strict to improve outcomes. However, this is a challenging task for patients, since they need to have access to quality healthcare resources that facilitate optimal GFD adherence. Understanding the strengths and weaknesses of the GFD, tackling coexisting nutritional deficiencies, and dealing with complex situations, such as seronegative CeD or non-responsive CeD, all require the involvement of a multidisciplinary team. The short- and long-term follow-up of CeD patients should preferably be performed by a combined Gastroenterology and Nutrition service with well-defined quality standards and the multidisciplinary involvement of physicians, nurses, dietitians, and psychologists. Nutritional advice and counseling by an experienced dietitian can reduce the costs associated with long-term follow-up of CeD patients. Likewise, psychological interventions may be essential in specific scenarios where implementing and sustaining a lifelong GFD can cause a significant psychological burden for patients. This manuscript aims to provide guidelines to improve clinical practice in the follow-up and monitoring of CeD patients and provide information on the nutritional risks of an ill-advised GFD. Clinicians, biochemists, food technologists, dietitians, and psychologists with a global view of the disease have been involved in its writing.
Collapse
Affiliation(s)
- Edurne Simón
- GLUTEN3S Research Group, Department of Nutrition and Food Science, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
| | - Marta Molero-Luis
- Laboratory of Gastroenterology and Trace Elements, Department of Laboratory Medicine, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Ricardo Fueyo-Díaz
- PROSAM Research Group (S69-23R), Department of Psychology and Sociology, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Cristian Costas-Batlle
- Department of Nutrition and Dietetics, Bradford Teaching Hospitals NHS Foundation Trust, Bradford BD9 6DA, UK
| | - Paula Crespo-Escobar
- ADViSE Research Group, Department of Health Science, European University Miguel de Cervantes, 47012 Valladolid, Spain
- Department of Nutrition and Obesity, Hospital Recoletas Campo Grande, 47007 Valladolid, Spain
| | - Miguel A Montoro-Huguet
- Gastroenterology, Hepatology and Nutrition Unit, University Hospital San Jorge, 22004 Huesca, Spain
- Department of Medicine, Faculty of Health and Sport Sciences, University of Zaragoza, 22002 Huesca, Spain
- Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| |
Collapse
|
45
|
Shen T, Wang H, Hu R, Lv Y. Developing neural network diagnostic models and potential drugs based on novel identified immune-related biomarkers for celiac disease. Hum Genomics 2023; 17:76. [PMID: 37587523 PMCID: PMC10433645 DOI: 10.1186/s40246-023-00526-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND As one of the most common intestinal inflammatory diseases, celiac disease (CD) is typically characterized by an autoimmune disorder resulting from ingesting gluten proteins. Although the incidence and prevalence of CD have increased over time, the diagnostic methods and treatment options are still limited. Therefore, it is urgent to investigate the potential biomarkers and targeted drugs for CD. METHODS Gene expression data was downloaded from GEO datasets. Differential gene expression analysis was performed to identify the dysregulated immune-related genes. Multiple machine algorithms, including randomForest, SVM-RFE, and LASSO, were used to select the hub immune-related genes (HIGs). The immune-related genes score (IG score) and artificial neural network (ANN) were constructed based on HIGs. Potential drugs targeting HIGs were identified by using the Enrichr platform and molecular docking method. RESULTS We identified the dysregulated immune-related genes at a genome-wide level and demonstrated their roles in CD-related immune pathways. The hub genes (MR1, CCL25, and TNFSF13B) were further screened by integrating several machine algorithms. Meanwhile, the CD patients were divided into distinct subtypes with either high- or low-immunoactivity using single-sample gene set enrichment analysis (ssGSEA) and consensus clustering. By constructing IG score based on HIGs, we found that patients with high IG score were mainly attributed to high-immunoactivity subgroups, which suggested a strong link between HIGs and immunoactivity of CD patients. In addition, the novel constructed ANN model showed the sound diagnostic ability of HIGs. Mechanistically, we validated that the HIGs play pivotal roles in regulating CD's immune and inflammatory state. Through targeting the HIGs, we also found potential drugs for anti-CD treatment by using the Enrichr platform and molecular docking method. CONCLUSIONS This study unveils the HIGs and elucidates the networks regulated by these genes in the context of CD. It underscores the pivotal significance of HIGs in accurately predicting the presence or absence of CD in patients. Consequently, this research offers promising prospects for the development of diagnostic biomarkers and therapeutic targets for CD.
Collapse
Affiliation(s)
- Tao Shen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Key Laboratory of Biomedicine in Gene Diseases, Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China.
| | - Haiyang Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Key Laboratory of Biomedicine in Gene Diseases, Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Rongkang Hu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Key Laboratory of Biomedicine in Gene Diseases, Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yanni Lv
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Key Laboratory of Biomedicine in Gene Diseases, Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
46
|
Guz-Mark A, Perets TT, Biran N, Jack Y, Zevit N, Silbermintz A, Matar M, Nachmias-Friedler V, Waisbourd-Zinman O, Bar-Lev MR, Huta Y, Ashorov O, Gingold-Belfer R, Shamir R. Gluten Immunogenic Peptides Are Not Correlated With Reported Adherence to Gluten-Free Diet in Children With Celiac Disease. J Pediatr Gastroenterol Nutr 2023; 77:244-248. [PMID: 37204826 DOI: 10.1097/mpg.0000000000003835] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
OBJECTIVE There is no gold standard to assess adherence to gluten-free diet (GFD) among patients with celiac disease (CeD). Gluten immunogenic peptides (GIPs) in urine and stool were suggested as novel markers for evaluating adherence to GFD. Our aim was to assess the presence of GIP in pediatric patients with CeD, and to compare the results with alternative methods for evaluating GFD adherence. METHODS Pediatric patients diagnosed with CeD, who were on GFD for at least 1 year, were enrolled and followed prospectively between November 2018 and January 2021. Study visits included clinical assessment, a dietitian interview, Biagi score, food questionnaires, anthropometric and laboratory measurements, and urine and stool samples obtained for laboratory GIP analysis. RESULTS The study included 74 patients (63.5% females), with median (interquartile range, IQR) age of 9.9 (7.8-11.7) years, and median (IQR) duration on GFD of 2.5 (2-5.5) years. Good GFD adherence, assessed by Biagi score, was reported in 93.1% of cases. GIP was evaluated during 134 visits, with GIP detected in 27 of 134 (20.1%) of the visits (16.3% of stool samples and 5.3% of urine samples). Positive GIP results were significantly more common in males compared to females (30.6% vs 14.1%, respectively, P < 0.05). Detection of positive GIP was not associated with dietary assessment of GFD adherence, celiac serology results, or reported symptoms. CONCLUSIONS Stool and urine GIP can be detected in children with CeD, even when dietary assessment indicate good adherence to GFD. The role of GIP testing in clinical practice should be further explored.
Collapse
Affiliation(s)
- Anat Guz-Mark
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tsachi Tsadok Perets
- the Gastroenterology Laboratory - Rabin Medical Center, Petach Tikva, Israel
- the Department of Digital Medical Technologies, Holon Institute of Technology, Holon, Israel
| | - Neta Biran
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Nutrition and Dietetics Department, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yifat Jack
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Nutrition and Dietetics Department, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Noam Zevit
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ari Silbermintz
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Manar Matar
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vered Nachmias-Friedler
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orith Waisbourd-Zinman
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Rozenfeld Bar-Lev
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yair Huta
- the Gastroenterology Laboratory - Rabin Medical Center, Petach Tikva, Israel
| | - Olga Ashorov
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- the Gastroenterology Laboratory - Rabin Medical Center, Petach Tikva, Israel
| | - Rachel Gingold-Belfer
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- the Nutrition and Dietetics Department, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Raanan Shamir
- From Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
47
|
Horton M, Olshan KL, Gleeson E, Regis S, Morson T, Hintze ZJ, Leonard MM, Silvester JA. Low Levels of Detectable Urine and Stool GIPs in Children with Celiac Disease on a Gluten-Free Diet. JPGN REPORTS 2023; 4:e323. [PMID: 37600614 PMCID: PMC10435043 DOI: 10.1097/pg9.0000000000000323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/05/2023] [Indexed: 08/22/2023]
Abstract
Objectives This study examines the prevalence of detectable gluten immunogenic peptides (GIPs) as a proxy for gluten exposure in children with celiac disease on a gluten-free diet in the United States, as estimated by gluten breakdown products excreted in urine and stool. Methods Urine and stool samples were collected in 3 settings (home, gastroenterology clinic, and endoscopy) for pediatric participants (ages 6-21 years old) across 2 medical centers. Commercial ELISA assays were used to quantify the GIPs in each sample. Results GIPs were detected in 4 out of 44 (9.1%) of stool samples and 6 out of 125 (4.8%) of urine samples provided by 84 children. These samples were collected across all settings, and most participants (70%) were asymptomatic at the time of sample collection. For the urine samples collected at the time of endoscopy, all subjects found to have persistent enteropathy had no detectable GIPs (0/12). Discussion GIPs provide an additional method for screening for gluten exposures in individuals with celiac disease on a gluten-free diet, and may be used across multiple settings. We found a low detection rate of GIPs in children. Our finding of undetectable GIPs in individuals with persistent enteropathy may be expected of a single determination under close observation or represent a lack of gluten exposure within the detection window. More research is needed to understand the dynamics of gluten absorption and excretion in the US pediatric population.
Collapse
Affiliation(s)
- Maxwell Horton
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine L. Olshan
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Elizabeth Gleeson
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Stephanie Regis
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor Morson
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Zackary J. Hintze
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maureen M. Leonard
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Jocelyn A. Silvester
- From the Celiac Research Program, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Nelson-Mora J, Rubio D, Ventura-Martínez A, González LA, Del-Rio D, Aranda-López Y, Jiménez-Díaz E, Zamarrón-Hernández D, Ríos-López DG, Aguirre S, Ruiz-Hernandez Y, Cruz-Ramírez A, Barjau JS, Jáurez MA, Lopez-Aparicio J, Campa-Higareda A, Fiordelisio T. New detection method of SARS-CoV-2 antibodies toward a point-of-care biosensor. Front Bioeng Biotechnol 2023; 11:1202126. [PMID: 37485316 PMCID: PMC10359622 DOI: 10.3389/fbioe.2023.1202126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The outbreak of COVID-19, a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, is regarded as the most severe of the documented coronavirus pandemics. The measurement and monitoring of SARS-CoV-2 antibody levels by serological tests are relevant for a better epidemiological and clinical understanding of COVID-19. The aim of this work was to design a method called the SARS-CoV-2 antibody detection method (SARS-CoV-2 AbDM) for fluorescence immunodetection of anti-SARS-CoV-2 IgG and IgM on both plate and microfluidic chip. For this purpose, a system with magnetic beads that immobilize the antigen (S protein and RBD) on its surface was used to determine the presence and quantity of antibodies in a sample in a single reaction. The SARS-CoV-2 AbDM led to several advantages in the performance of the tests, such as reduced cost, possibility of performing isolated or multiple samples, potential of multiplex detection, and capacity to detect whole blood samples without losing resolution. In addition, due to the microfluidic chip in conjunction with the motorized actuated platform, the time, sample quantity, and operator intervention during the process were reduced. All these advantages suggest that the SARS-CoV-2 AbDM has the potential to be developed as a PoC that can be used as a tool for seroprevalence monitoring, allowing a better understanding of the epidemiological and clinical characteristics of COVID-19 and contributing to more effective and ethical decision-making in strategies to fight against the COVID-19 pandemic.
Collapse
Affiliation(s)
- Janikua Nelson-Mora
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Rubio
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Amairani Ventura-Martínez
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis A. González
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Del-Rio
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Neuroendocrinología Comparada-LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yuli Aranda-López
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar Jiménez-Díaz
- Laboratorio de Neuroendocrinología Comparada-LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Imagenología Cuantitativa, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diego Zamarrón-Hernández
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana G. Ríos-López
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephanie Aguirre
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yasab Ruiz-Hernandez
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aarón Cruz-Ramírez
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jonás S. Barjau
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel A. Jáurez
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jehú Lopez-Aparicio
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Andrea Campa-Higareda
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Tatiana Fiordelisio
- Unidad de Biología Molecular y Diagnóstico, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Neuroendocrinología Comparada-LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Imagenología Cuantitativa, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
49
|
Monachesi C, Catassi G, Catassi C. The use of urine peptidomics to define dietary gluten peptides from patients with celiac disease and the clinical relevance. Expert Rev Proteomics 2023; 20:281-290. [PMID: 37855116 DOI: 10.1080/14789450.2023.2270775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION Determination of urinary gluten immunogenic peptides (GIP) has emerged as one of the most attractive test to monitor the adherence to the gluten-free diet (GFD) of patients with celiac disease (CD), being a simple, noninvasive and direct method to detect gluten contamination of the GFD. AREAS COVERED We conducted a scoping review in Medline (PubMed) of articles published up to April 2023 that analyzed any aspect of the clinical relevance of the use of urinary GIP measurement in patients with CD. A total of 17 articles reporting the clinical use of urinary peptidomics for the follow-up of CD patients were finally included. EXPERT OPINION Available data suggest that a negative urinary GIP result is a reliable noninvasive predictor of intestinal mucosa healing in CD patients treated with the GFD, especially if testing three urine samples on different days including the weekend. Due to conflicting results about the sensitivity and the specificity of the urinary GIP determination, additional in-depth information is needed, particularly related to (1) the relationship between the amount of ingested gluten and the quantity of urinary GIP excreted in treated CD patients, (2) the GIP kinetics and best timing for sample collection.
Collapse
Affiliation(s)
- Chiara Monachesi
- Division of Pediatrics and Center for Celiac Research, DISCO Department, Marche Polytechnic University, Ancona, Italy
| | - Giulia Catassi
- Pediatric Gastroenterology and Liver Unit, Department of Maternal and Child Health, Sapienza-University of Rome, Rome, Italy
| | - Carlo Catassi
- Division of Pediatrics and Center for Celiac Research, DISCO Department, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
50
|
Engel S, Klotz L, Wirth T, Fleck AK, Pickert G, Eschborn M, Kreuzburg S, Curella V, Bittner S, Zipp F, Schuppan D, Luessi F. Attenuation of immune activation in patients with multiple sclerosis on a wheat-reduced diet: a pilot crossover trial. Ther Adv Neurol Disord 2023; 16:17562864231170928. [PMID: 37384112 PMCID: PMC10293514 DOI: 10.1177/17562864231170928] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/03/2023] [Indexed: 06/30/2023] Open
Abstract
Background Western lifestyle has been associated with an increase in relapsing-remitting multiple sclerosis (RRMS). In mice, dietary wheat amylase-trypsin inhibitors (ATIs) activate intestinal myeloid cells and augment T cell-mediated systemic inflammation. Objective The aim of this study was to assess whether a wheat- and thus ATI-reduced diet might exert beneficial effects in RRMS patients with modest disease activity. Methods In this 6-month, crossover, open-label, bicentric proof-of-concept trial, 16 RRMS patients with stable disease course were randomized to either 3 months of a standard wheat-containing diet with consecutive switch to a > 90% wheat-reduced diet, or vice versa. Results The primary endpoint was negative, as the frequency of circulating pro-inflammatory T cells did not decrease during the ATI-reduced diet. We did, however, observe decreased frequencies of CD14+ CD16++ monocytes and a concomitant increase in CD14++ CD16- monocytes during the wheat-reduced diet interval. This was accompanied by an improvement in pain-related quality of life in health-related quality of life assessed (SF-36). Conclusion Our results suggest that the wheat- and thus ATI-reduced diet was associated with changes in monocyte subsets and improved pain-related quality of life in RRMS patients. Thus, a wheat (ATI)-reduced diet might be a complementary approach accompanying immunotherapy for some patients. Registration German Clinical Trial Register (No. DRKS00027967).
Collapse
Affiliation(s)
- Sinah Engel
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Timo Wirth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Ann-Katrin Fleck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Geethanjali Pickert
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Eschborn
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Samia Kreuzburg
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Valentina Curella
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Felix Luessi
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| |
Collapse
|