1
|
Lenti MV, Bianchi PI, Di Sabatino A. Coeliac disease and chronic liver disease: a double-face issue. THE LANCET REGIONAL HEALTH. EUROPE 2025; 50:101216. [PMID: 39925796 PMCID: PMC11804579 DOI: 10.1016/j.lanepe.2025.101216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Italy
- Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | | | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Italy
- Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
2
|
Scarampi M, Mengoli C, Miceli E, Di Stefano M. Vitamins and Celiac Disease: Beyond Vitamin D. Metabolites 2025; 15:78. [PMID: 39997703 PMCID: PMC11857425 DOI: 10.3390/metabo15020078] [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: 12/16/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Celiac disease is a chronic inflammatory condition of the small bowel caused, in genetically predisposed subjects, by the ingestion of gluten and characterised by a broad clinical polymorphism, ranging from patients with an asymptomatic or paucisymptomatic disease. The clinical presentation ranges from the presence of minor, apparently unrelated symptoms or first-degree kinship with known patients to severe intestinal malabsorption and all its clinical consequences and complications. Even if a large body of research improved our understanding of the molecular basis of celiac disease pathophysiology, enhancing the identification of new targets for future new treatments, an accurate gluten-free diet remains the mainstay of the therapy for this condition, restoring a normal absorptive mucosa. It is very rare, nowadays, to deal with patients with severe malabsorption syndrome secondary to celiac disease. Consequently, physicians are currently less prone to search for nutritional deficiencies in celiac disease. To pinpoint the possibility of both a disease-related and a diet-induced vitamin deficiency, we reviewed the literature on vitamin deficiency in this condition and reported the impact both in untreated and treated patients with celiac disease. A gluten-free diet must be tailored for each patient to meet nutritional targets: the pre-existence or diet-induced intake inadequacies should be carefully considered for an effective management of celiac disease.
Collapse
Affiliation(s)
| | | | | | - Michele Di Stefano
- 1st Department of Medicine, IRCCS “S.Matteo” Hospital Foundation, 27100 Pavia, Italy; (M.S.); (C.M.); (E.M.)
| |
Collapse
|
3
|
Seidita A, Latteri F, Pistone M, Giuliano A, Bertoncello L, Cavallo G, Chiavetta M, Faraci F, Nigro A, Termini A, Verona L, Ammannato A, Accomando S, Cavataio F, Lospalluti ML, Citrano M, Di Liberto D, Soresi M, Mansueto P, Carroccio A. Celiac Disease and Liver Damage: The Gut-Liver Axis Strikes Back (Again)? A Retrospective Analysis in the Light of a Literature Review. Nutrients 2024; 17:85. [PMID: 39796519 PMCID: PMC11722968 DOI: 10.3390/nu17010085] [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: 12/07/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: An increasing number of studies have reported liver involvement in both children and adults with celiac disease (CD). This often manifests as isolated hypertransaminasemia or hepatic steatosis (HS). The aim of this study was to define the prevalence of hypertransaminasemia and HS in a pediatric population with CD before starting a gluten-free diet (GFD) and to analyze how the introduction of a GFD could modify this condition. We also conducted a state-of-the-art literature review of the association between hypertransaminasemia, metabolic dysfunction-associated steatotic liver disease (MASLD) and CD. Methods: We retrospectively reviewed the clinical charts of pediatric CD patients diagnosed in three different pediatric units of Sicily, analyzing clinical, laboratory, ultrasound, and histology data before and 12 months after the introduction of a GFD. Results: A total of 160 patients (65.0% females, median age 6.4 (0.8-13.2) years) were included; hypertransaminasemia and HS prevalences at diagnosis were 8.1% and 6.1%, respectively. Subjects with hypertransaminasemia were younger (p = 0.01) than those without and had higher frequencies of HS (p = 0.034) and anti-tissue transglutaminase (tTg) immunoglobulin (Ig)G positivity (p = 0.046). Subjects with HS were younger (p = 0.0001) and had a higher frequency of hypertransaminasemia (p = 0.029) compared to non-steatotic ones. After 12 months of a GFD, hypertransaminasemia and HS persisted in 53.8% and 50.0% of patients, respectively. Conclusions: The prevalences of hypertransaminasemia and HS in Sicilian pediatric CD patients seem to be lower than those reported in other geographical areas. A GFD can reverse the trend of liver involvement, although periods of longer than 12 months may be necessary. However, a GFD has been associated with an increased prevalence of HS, and so regular follow-up involving a nutritionist should be recommended to guide physicians in patient management.
Collapse
Affiliation(s)
- Aurelio Seidita
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Federica Latteri
- Gastroenterology Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy
| | - Mirco Pistone
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Alessandra Giuliano
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Luca Bertoncello
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Giorgia Cavallo
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Marta Chiavetta
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Francesco Faraci
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Alessia Nigro
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Alessandro Termini
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Laura Verona
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Agnese Ammannato
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Salvatore Accomando
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
- Department of Pediatrics, University Hospital of Palermo, 90134 Palermo, Italy
| | - Francesca Cavataio
- Pediatric Gastroenterology Unit, “Di Cristina” Hospital, Palermo, 90134 Palermo, Italy
| | | | - Michele Citrano
- Pediatrics Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy
| | - Diana Di Liberto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
| | - Maurizio Soresi
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Pasquale Mansueto
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| | - Antonio Carroccio
- Internal Medicine Unit, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, Via Trabucco, 180, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche, 2, 90127 Palermo, Italy
| |
Collapse
|
4
|
Marchi E, Craig JW, Kalac M. Current and upcoming treatment approaches to uncommon subtypes of PTCL (EATL, MEITL, SPTCL, and HSTCL). Blood 2024; 144:1898-1909. [PMID: 38657272 DOI: 10.1182/blood.2023021788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
ABSTRACT Rare subtypes of peripheral T-cell lymphoma (PTCL) including enteropathy-associated T-cell lymphoma (EATL), monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL), subcutaneous panniculitis-like T-cell lymphoma (SPTCL), and hepatosplenic T-cell lymphoma (HSTCL) are underrepresented in most registries and clinical studies. Most of the literature is obtained from small case series, single-institution retrospective studies, and subgroup analyses of the largest studies with few recent and ongoing exceptions. Although the pathogenesis and biology of these entities have yet to be fully elucidated, global efforts by the scientific community have started to shed some light on the most frequently deregulated pathways. In this review, we highlight the most pertinent clinical and pathologic features of rare subtypes of PTCL including EATL/MEITL, SPTCL, and HSTCL. We also summarize the results of recent developments identifying potential targets for novel therapeutic strategies based on molecular studies. Finally, we highlight the underrepresentation of these rare subtypes in most clinical trials, making evidence-based therapeutic decisions extremely challenging.
Collapse
Affiliation(s)
- Enrica Marchi
- Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
| | - Jeffrey W Craig
- Department of Pathology, University of Virginia Cancer Center, Charlottesville, VA
| | - Matko Kalac
- Department of Hematology and Oncology, University of California, Irvine, CA
| |
Collapse
|
5
|
Urganci Ü. Celiac Disease and Gut Microbiota: Herbal Treatment and Gluten-Free Diet. HERBAL MEDICINE FOR AUTOIMMUNE DISEASES 2024:159-184. [DOI: 10.2174/9789815305005124010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Celiac disease (CD) manifests as a targeted autoimmune response that
adversely affects the small intestine, primarily affecting individuals with a particular
genetic predisposition. Diagnosis centers on identifying this gluten-sensitive
enteropathy, which can be ameliorated through the implementation of a gluten-free diet
(GFD), correlating with mucosal healing and symptom alleviation. The human
microbiota, a vast symbiotic community within the gastrointestinal tract, profoundly
impacts human health. Advances in genome sequencing have elucidated the intricate
relationship between gut microbiota and autoimmune diseases, including CD,
emphasizing the significant role of dietary patterns in shaping the gut microbiota. The
influence of GFD on microbiota composition, the only clinically validated treatment
for CD, leads to a nutritional shift and potential macronutrient imbalance. Emerging
research also highlights the therapeutic potential of various herbs with antioxidant,
anti-inflammatory, antimicrobial, gastroprotective, and immunomodulatory properties
as complementary approaches to manage CD. This chapter synthesizes the complex
interactions between genetics, diet, gut microbiota, and potential herbal interventions in
CD, paving the way for more comprehensive understanding and management
strategies.
Collapse
Affiliation(s)
- Ünkan Urganci
- Department of Food Engineering, Faculty of Engineering, Pamukkale University, Denizli 20160,
Türkiye
| |
Collapse
|
6
|
Kumari B, Tiwari A, Meena S, Ahirwar DK. Inflammation-Associated Stem Cells in Gastrointestinal Cancers: Their Utility as Prognostic Biomarkers and Therapeutic Targets. Cancers (Basel) 2024; 16:3134. [PMID: 39335106 PMCID: PMC11429849 DOI: 10.3390/cancers16183134] [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: 08/04/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Stem cells are critical for the development and homeostasis of the gastrointestinal (GI) tract. Inflammatory molecules are known to regulate the activity of stem cells. A comprehensive review specifically describing the role of inflammatory molecules in the regulation of stem cells within the GI tract and in GI cancers (GICs) is not available. This review focuses on understanding the role of inflammatory molecules and stem cells in maintaining homeostasis of the GI tract. We further discuss how inflammatory conditions contribute to the transformation of stem cells into tumor-initiating cells. We also describe the molecular mechanisms of inflammation and stem cell-driven progression and metastasis of GICs. Furthermore, we report on studies describing the prognostic value of cancer stem cells and the clinical trials evaluating their therapeutic utility. This review provides a detailed overview on the role of inflammatory molecules and stem cells in maintaining GI tract homeostasis and their implications for GI-related malignancies.
Collapse
Affiliation(s)
- Beauty Kumari
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342030, Rajasthan, India; (B.K.); (A.T.)
| | - Aniket Tiwari
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342030, Rajasthan, India; (B.K.); (A.T.)
| | - Sakshi Meena
- School of Life Sciences, Devi Ahilya Vishwavidyalaya Indore, Indore 452001, Madhya Pradesh, India;
| | - Dinesh Kumar Ahirwar
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342030, Rajasthan, India; (B.K.); (A.T.)
| |
Collapse
|
7
|
Stødle IH, Koldsland OC, Lukina P, Andersen IL, Mjønes P, Rønne E, Høvik H, Ness-Jensen E, Verket A. Undiagnosed Celiac Disease and Periodontal Bone Loss: A Cross-Sectional Radiological Assessment from the HUNT Study. Int J Dent 2024; 2024:1952244. [PMID: 39257416 PMCID: PMC11383648 DOI: 10.1155/2024/1952244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/27/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
Objective The objective was to assess radiographic periodontal bone loss in a population with previously undiagnosed celiac disease, and to compare it to a reference group without celiac disease. Background Periodontitis and celiac disease are chronic inflammatory diseases with possible similar features related to immune reactions and microbial dysbiosis. The relationship between these two diseases is not clear. Methods Clinical variables, blood samples, and answers to questionnaires were collected from participants in the fourth Trøndelag Health Study (HUNT4). Celiac disease was determined based on transglutaminase 2 (TG2), immunoglobulin A (IgA), and G (IgG) in serum samples. Seropositive individuals were invited to endoscopic examination and tissue sampling. Radiographically assessed bone loss caused by periodontitis in two different levels of severity was applied as outcome, that is, ≥15% and >33% of root length. Bone loss was determined in panoramic images in participants that had attended radiographic examination in the HUNT4 Oral Health Study or in the HUNT4 Coeliac Disease Study. The association between previously undiagnosed celiac disease and radiographic bone loss was estimated by adjusted Poisson regression models. Results Radiographic assessment was completed in 485 individuals with celiac disease determined by positive serology and in 4,727 individuals with negative serology (without celiac disease). Compared to nonceliacs, seropositive participants were less likely to present with ≥15% radiographic bone loss (prevalence ratio (PR) 0.89 (95% CI 0.84-0.96). A similar association was also observed after histopathological confirmation of celiac disease (PR 0.89 (95% CI 0.82-0.98). No association between undiagnosed celiac disease and periodontal bone loss was observed when analyses were limited to individuals with severe bone loss (>33%). Conclusion In this study of previously undiagnosed celiac disease and periodontal bone loss, newly diagnosed celiac disease was associated with less likelihood of presenting with ≥15% radiographic bone loss compared to a nonceliac reference group.
Collapse
Affiliation(s)
- Ida Haukåen Stødle
- Department of Periodontology Institute of Clinical Dentistry Faculty of Dentistry University of Oslo, Oslo, Norway
| | - Odd Carsten Koldsland
- Department of Periodontology Institute of Clinical Dentistry Faculty of Dentistry University of Oslo, Oslo, Norway
| | - Polina Lukina
- HUNT Research Centre Department of Public Health and Nursing Norwegian University of Science and Technology (NTNU), Levanger, Norway
| | - Ina L Andersen
- HUNT Research Centre Department of Public Health and Nursing Norwegian University of Science and Technology (NTNU), Levanger, Norway
- Department of Medicine Levanger Hospital Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Patricia Mjønes
- Department of Clinical and Molecular Medicine Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Pathology St. Olav's Hospital Trondheim University Hospital, Trondheim, Norway
| | - Elin Rønne
- Department of Pathology St. Olav's Hospital Trondheim University Hospital, Trondheim, Norway
| | - Hedda Høvik
- Center for Oral Health Services and Research, Mid-Norway (TkMidt), Trondheim, Norway
| | - Eivind Ness-Jensen
- HUNT Research Centre Department of Public Health and Nursing Norwegian University of Science and Technology (NTNU), Levanger, Norway
- Department of Medicine Levanger Hospital Nord-Trøndelag Hospital Trust, Levanger, Norway
- Department of Molecular Medicine and Surgery Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anders Verket
- Department of Periodontology Institute of Clinical Dentistry Faculty of Dentistry University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Rigo FF, Oliveira ECSD, Quaglio AEV, Moutinho BD, Di Stasi LC, Sassaki LY. Expression of MicroRNAs in Adults with Celiac Disease: A Narrative Review. Int J Mol Sci 2024; 25:9412. [PMID: 39273359 PMCID: PMC11395070 DOI: 10.3390/ijms25179412] [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: 07/06/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by the ingestion of proline- and glutamine-rich proteins, widely termed "gluten", in genetically susceptible individuals. CD induces an altered immune response that leads to chronic inflammation and duodenal mucosal damage. Currently, there are no specific tests for the accurate diagnosis of CD, and no drugs are available to treat this condition. The only available treatment strategy is lifelong adherence to a gluten-free diet. However, some studies have investigated the involvement of microRNAs (miRNAs) in CD pathogenesis. miRNAs are small noncoding ribonucleic acid molecules that regulate gene expression. Despite the growing number of studies on the role of miRNAs in autoimmune disorders, data on miRNAs and CD are scarce. Therefore, this study aimed to perform a literature review to summarize CD, miRNAs, and the potential interactions between miRNAs and CD in adults. This review shows that miRNA expression can suppress or stimulate pathways related to CD pathogenesis by regulating cell proliferation and differentiation, regulatory T-cell development, innate immune response, activation of the inflammatory cascade, focal adhesion, T-cell commitment, tissue transglutaminase synthesis, and cell cycle. Thus, identifying miRNAs and their related effects on CD could open new possibilities for diagnosis, prognosis, and follow-up of biomarkers.
Collapse
Affiliation(s)
- Francielen Furieri Rigo
- Department of Internal Medicine, Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | | | | | - Bruna Damásio Moutinho
- Department of Gastroenterology, Division of Clinical Gastroenterology and Hepatology, University of São Paulo School of Medicine (USP), São Paulo 01246-903, SP, Brazil
| | - Luiz Claudio Di Stasi
- Laboratory of Phytomedicines, Pharmacology and Biotechnology (PhytoPharmaTec), Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Ligia Yukie Sassaki
- Department of Internal Medicine, Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
9
|
Siddiqui MS, Shahi MH, Castresana JS. The role of the adenylate kinase 5 gene in various diseases and cancer. J Clin Transl Sci 2024; 8:e96. [PMID: 39655021 PMCID: PMC11626602 DOI: 10.1017/cts.2024.536] [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: 11/10/2023] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024] Open
Abstract
Adenylate kinases (AKs) are important enzymes involved in cellular energy metabolism. Among AKs, AK5 (adenylate kinase 5), a cytosolic protein, is emerging as a significant contributor to various diseases and cellular processes. This comprehensive review integrates findings from various research groups on AK5 since its discovery, shedding light on its multifaceted roles in nucleotide metabolism, energy regulation, and cellular differentiation. We investigate its implications in a spectrum of diseases, including autoimmune encephalitis, epilepsy, neurodegenerative disorders such as Alzheimer's and Parkinson's, diabetes, lower extremity arterial disease, celiac disease, and various cancers. Notably, AK5's expression levels and methylation status have been associated with cancer progression and patient outcomes, indicating its potential as a prognostic indicator. Furthermore, AK5 is implicated in regulating cellular processes in breast cancer, gastric cancer, colorectal carcinoma, prostate cancer, and colon adenocarcinoma, suggesting its relevance across different cancer types. However, a limitation lies in the need for more robust clinical validation and a deeper understanding of AK5's precise mechanisms in disease pathogenesis, despite its association with various pathophysiological conditions. Nonetheless, AK5 holds promise as a therapeutic target, with emerging evidence suggesting its potential in therapy development.
Collapse
Affiliation(s)
- M. Sarim Siddiqui
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh202002, India
| | - Mehdi H. Shahi
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh202002, India
| | - Javier S. Castresana
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona31008, Spain
| |
Collapse
|
10
|
Zheng W, Pang K, Min Y, Wu D. Prospect and Challenges of Volatile Organic Compound Breath Testing in Non-Cancer Gastrointestinal Disorders. Biomedicines 2024; 12:1815. [PMID: 39200279 PMCID: PMC11351786 DOI: 10.3390/biomedicines12081815] [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: 06/16/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Breath analysis, despite being an overlooked biomatrix, has a rich history in disease diagnosis. However, volatile organic compounds (VOCs) have yet to establish themselves as clinically validated biomarkers for specific diseases. As focusing solely on late-stage or malignant disease biomarkers may have limited relevance in clinical practice, the objective of this review is to explore the potential of VOC breath tests for the diagnosis of non-cancer diseases: (1) Precancerous conditions like gastro-esophageal reflux disease (GERD) and Barrett's esophagus (BE), where breath tests can complement endoscopic screening; (2) endoluminal diseases associated with autoinflammation and dysbiosis, such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and coeliac disease, which currently rely on biopsy and symptom-based diagnosis; (3) chronic liver diseases like cirrhosis, hepatic encephalopathy, and non-alcoholic fatty liver disease, which lack non-invasive diagnostic tools for disease progression monitoring and prognostic assessment. A literature search was conducted through EMBASE, MEDLINE, and Cochrane databases, leading to an overview of 24 studies. The characteristics of these studies, including analytical platforms, disorder type and stage, group size, and performance evaluation parameters for diagnostic tests are discussed. Furthermore, how VOCs can be utilized as non-invasive diagnostic tools to complement existing gold standards is explored. By refining study designs, sampling procedures, and comparing VOCs in urine and blood, we can gain a deeper understanding of the metabolic pathways underlying VOCs. This will establish breath analysis as an effective non-invasive method for differential diagnosis and disease monitoring.
Collapse
Affiliation(s)
- Weiyang Zheng
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China;
| | - Ke Pang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100006, China; (K.P.); (Y.M.)
| | - Yiyang Min
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100006, China; (K.P.); (Y.M.)
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China;
- Clinical Epidemiology Unit, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
11
|
Ghorbanzadeh S, Khojini JY, Abouali R, Alimardan S, Zahedi M, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Clearing the Path: Exploring Apoptotic Cell Clearance in Inflammatory and Autoimmune Disorders for Therapeutic Advancements. Mol Biotechnol 2024:10.1007/s12033-024-01222-6. [PMID: 38935260 DOI: 10.1007/s12033-024-01222-6] [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: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 06/28/2024]
Abstract
Inflammatory and autoimmune disorders, characterized by dysregulated immune responses leading to tissue damage and chronic inflammation, present significant health challenges. This review uniquely focuses on efferocytosis-the phagocyte-mediated clearance of apoptotic cells-and its pivotal role in these disorders. We delve into the intricate mechanisms of efferocytosis' four stages and their implications in disease pathogenesis, distinguishing our study from previous literature. Our findings highlight impaired efferocytosis in conditions like atherosclerosis and asthma, proposing its targeting as a novel therapeutic strategy. We discuss the therapeutic potential of efferocytosis in modulating immune responses and resolving inflammation, offering a new perspective in treating inflammatory disorders.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran.
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
12
|
Macedo L, Catarino M, Festas C, Alves P. Vulnerability in Children with Celiac Disease: Findings from a Scoping Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:729. [PMID: 38929308 PMCID: PMC11201835 DOI: 10.3390/children11060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
(1) Background: The scientific literature highlights that children diagnosed with celiac disease (CD) are at a heightened risk of experiencing physical, psychological, and social challenges, impacting their overall healthy childhood development. However, there remains a lack of a clear understanding regarding the factors that contribute to this vulnerability. The purpose of this study is to analyze and map the evidence on the sociopsychosomatic vulnerability of these children and identify gaps in this topic. (2) Methods: Following Joanna Briggs Institute's guidelines for scoping reviews, we executed a detailed search of key electronic databases and explored the grey literature to capture a broad spectrum of studies. Our focus was on identifying research that looked into the multiple dimensions of vulnerability-physical, psychological, and social-in children with CD. We included a diverse range of study designs as well as systematic reviews, ensuring a comprehensive analysis. The selection process was stringent, utilizing clearly defined inclusion and exclusion criteria. (3) Results: We identified 61 studies that met our inclusion criteria. The review highlighted significant adverse health outcomes in children with CD and elucidated various individual and environmental determinants that influenced these vulnerabilities. It also underscored the lack of assessment tools to evaluate the risk of health problems in this population. (4) Conclusions: The findings underscore a critical need for further research to deepen our understanding of the vulnerabilities associated with CD in children. Developing targeted assessment tools will be crucial in stratifying health risks and enhancing care strategies for this vulnerable population.
Collapse
Affiliation(s)
- Lúcia Macedo
- Âncora Community Care Unit, Gaia and Espinho Local Health Unit, 4430-037 Gaia, Portugal
- Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (C.F.); (P.A.)
- Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal;
| | - Marta Catarino
- Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal;
- Health Department, Polytechnic Institute of Beja, 7800-111 Beja, Portugal
- Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 1649-023 Lisboa, Portugal
| | - Constança Festas
- Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (C.F.); (P.A.)
- Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal;
| | - Paulo Alves
- Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (C.F.); (P.A.)
- Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS), Universidade Católica Portuguesa, 4169-005 Porto, Portugal;
| |
Collapse
|
13
|
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
|
14
|
Velikova T, Vasilev G, Shumnalieva R, Chervenkov L, Miteva DG, Gulinac M, Priftis S, Lazova S. Autoantibodies related to ataxia and other central nervous system manifestations of gluten enteropathy. World J Clin Cases 2024; 12:2031-2039. [PMID: 38680259 PMCID: PMC11045506 DOI: 10.12998/wjcc.v12.i12.2031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/07/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Gluten ataxia and other central nervous system disorders could be linked to gluten enteropathy and related autoantibodies. In this narrative review, we focus on the various neuro-logical manifestations in patients with gluten sensitivity/celiac disease, immunological and autoimmune mechanisms of ataxia in connection to gluten sensitivity and the autoantibodies that could be used as a biomarker for diagnosing and following. We focused on the anti-gliadin antibodies, antibodies to different isoforms of tissue transglutaminase (TG) (anti-TG2, 3, and 6 antibodies), anti-glycine receptor antibodies, anti-glutamine acid decarboxylase antibodies, anti-deamidated gliadin peptides antibodies, etc. Most studies found a higher prevalence of these antibodies in patients with gluten sensitivity and neurological dysfunction, presented as different neurological disorders. We also discuss the role of a gluten-free diet on the clinical improvement of patients and also on imaging of these disorders.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
| | - Georgi Vasilev
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
- Clinic of Neurology and Department of Emergency Medicine, UMHAT "Sv. Georgi", Plovdiv 4000, Bulgaria
| | - Russka Shumnalieva
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
- Department of Rheumatology, Clinic of Rheumatology, University Hospital "St. Ivan Rilski", Medical University-Sofia, Sofia 1612, Bulgaria
| | - Lyubomir Chervenkov
- Department of Diagnostic Imaging, Medical University Plovdiv, Plovdiv 4000, Bulgaria
| | - Dimitrina Georgieva Miteva
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
- Department of Genetics, Faculty of Biology, Sofia University "St. Kliment Ohridski", Sofia 1164, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Plovdiv 4002, Bulgaria
| | - Stamatios Priftis
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Sofia 1407, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University "St. Kliment Ohridski", Sofia 1407, Bulgaria
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Sofia 1407, Bulgaria
- Department of Pediatric, University Hospital "N. I. Pirogov", Sofia 1606, Bulgaria
| |
Collapse
|
15
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [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: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
16
|
Yadav M, Chauhan NS. Role of gut-microbiota in disease severity and clinical outcomes. Brief Funct Genomics 2024; 23:24-37. [PMID: 36281758 DOI: 10.1093/bfgp/elac037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 01/21/2024] Open
Abstract
A delicate balance of nutrients, antigens, metabolites and xenobiotics in body fluids, primarily managed by diet and host metabolism, governs human health. Human gut microbiota is a gatekeeper to nutrient bioavailability, pathogens exposure and xenobiotic metabolism. Human gut microbiota starts establishing during birth and evolves into a resilient structure by adolescence. It supplements the host's metabolic machinery and assists in many physiological processes to ensure health. Biotic and abiotic stressors could induce dysbiosis in gut microbiota composition leading to disease manifestations. Despite tremendous scientific advancements, a clear understanding of the involvement of gut microbiota dysbiosis during disease onset and clinical outcomes is still awaited. This would be important for developing an effective and sustainable therapeutic intervention. This review synthesizes the present scientific knowledge to present a comprehensive picture of the role of gut microbiota in the onset and severity of a disease.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
17
|
Lenti MV, Broglio G, Lucioni M, Corazza GR. Refractory celiac disease and lymphomagenesis. PEDIATRIC AND ADULT CELIAC DISEASE 2024:207-227. [DOI: 10.1016/b978-0-443-13359-6.00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
18
|
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]
|
19
|
Ancona S, Bianchin S, Zampatti N, Nosratian V, Bigatti C, Ferro J, Trambaiolo Antonelli C, Viglizzo G, Gandullia P, Malerba F, Crocco M. Cutaneous Disorders Masking Celiac Disease: Case Report and Mini Review with Proposal for a Practical Clinical Approach. Nutrients 2023; 16:83. [PMID: 38201912 PMCID: PMC10780572 DOI: 10.3390/nu16010083] [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: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Celiac disease (CD) is an immune-mediated systemic gluten-related disorder characterized by a wide spectrum of intestinal and extra-intestinal manifestations, including damage to cutaneous and connective tissue. We report a rare case of chronic severe dermatitis involving connective tissue and cutaneous vascular vessels as the main clinical presentation of undiagnosed seronegative gluten disorder. A gluten-free diet dramatically improved the intestinal and cutaneous clinical damage in the patient. Pitfalls and the steps of differential diagnosis are described. We also review the literature regarding studies of CD and connective tissue diseases to extend the knowledge of these rare associations. We propose a practical diagnostic approach in suspected CD in autoimmune cutaneous disorders.
Collapse
Affiliation(s)
- Silvana Ancona
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy (P.G.); (F.M.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, 16132 Genoa, Italy
| | - Silvia Bianchin
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, 16132 Genoa, Italy
| | - Noemi Zampatti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, 16132 Genoa, Italy
| | | | - Carolina Bigatti
- UO Nephrology Dialysis and Transplant, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Jacopo Ferro
- Pathology Unit, U.O.C. Anatomia Patologica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (J.F.); (C.T.A.)
| | - Chiara Trambaiolo Antonelli
- Pathology Unit, U.O.C. Anatomia Patologica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (J.F.); (C.T.A.)
| | | | - Paolo Gandullia
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy (P.G.); (F.M.)
| | - Federica Malerba
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy (P.G.); (F.M.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, 16132 Genoa, Italy
| | - Marco Crocco
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy (P.G.); (F.M.)
| |
Collapse
|
20
|
Franco L, Nakano EY, Raposo A, Alturki HA, Alarifi SN, Chaves C, Teixeira-Lemos E, Romão B. Eating Attitudes of Patients with Celiac Disease in Brazil: A Nationwide Assessment with the EAT-26 Instrument. Nutrients 2023; 15:4796. [PMID: 38004190 PMCID: PMC10674570 DOI: 10.3390/nu15224796] [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: 10/21/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by the ingestion of gluten in genetically predisposed individuals. In this sense, a gluten-free diet is the only safe treatment available. Due to the restrictions resulting from this eating pattern, this treatment may impair the relationship of the people with CD with food, increasing the risk of a disordered eating attitude, which is associated with eating disorders. The EAT-26 is a validated instrument already applied worldwide in different populations, and higher scores are suggestive of eating attitudes prone to evolve into eating disorders. Studies carried out in other countries have already shown that people with CD are prone to developing eating disorders; however, no study has been carried out with this theme in the population with CD in Brazil. We carried out a nationwide cross-sectional study in three steps: (i) study design and instrument; (ii) recruitment of participants and ethics; (iii) statistical analysis. A total of 385 participants were included in our sample, 96.36% of them being women. The internal consistency of the applied self-administered Brazilian version of the EAT-26 online questionnaire presented a satisfactory Cronbach's alpha of 0.812, and in total, 36.1% of the respondents were classified with a disordered eating attitude. No differences were found among the scores of participants when divided by categories regarding gender, average monthly income, age, and educational level. However, scores classified as a disordered eating attitude were found in respondents with a body mass index classified as overweight and obese. Our study highlights that disordered eating attitudes are present in overweight and obese women with celiac disease; thus, public health politics are needed to prevent and treat these attitudes.
Collapse
Affiliation(s)
- Luiza Franco
- Instituto de Educação Superior de Brasilia, IESB University Center, Brasília 70200-730, Brazil;
| | | | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Hmidan A. Alturki
- General Directorate for Funds & Grants, King Abdulaziz City for Science & Technology, Riyadh 11442, Saudi Arabia;
| | - Sehad N. Alarifi
- Department of Food and Nutrition Science, Al-Quwayiyah College of Sciences and Humanities, Shaqra University, Shaqraa 11971, Saudi Arabia;
| | - Cláudia Chaves
- ESSV, Centre for Studies in Education and Innovation (CI&DEI), Polytechnic University of Viseu, 3504-510 Viseu, Portugal;
| | - Edite Teixeira-Lemos
- CERNAS Research Centre, Polytechnic University of Viseu, 3504-510 Viseu, Portugal;
| | - Bernardo Romão
- Instituto de Educação Superior de Brasilia, IESB University Center, Brasília 70200-730, Brazil;
| |
Collapse
|
21
|
Mousavi Maleki MS, Aghamirza Moghim Ali Abadi H, Vaziri B, Shabani AA, Ghavami G, Madanchi H, Sardari S. Bromelain and ficin proteolytic effects on gliadin cytotoxicity and expression of genes involved in cell-tight junctions in Caco-2 cells. Amino Acids 2023; 55:1601-1619. [PMID: 37803248 DOI: 10.1007/s00726-023-03333-x] [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: 07/26/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023]
Abstract
Enzyme therapy for celiac disease (CeD), which digests gliadin into non-immunogenic and non-toxic peptides, can be an appropriate treatment option for CeD. Here, we have investigated the effectiveness of bromelain and ficin on gliadin digestion using in vitro, such as SDS-PAGE, HPLC, and circular dichroism (CD). Furthermore, the cytotoxicity of gliadin and 19-mer peptide before and after digestion with these enzymes was evaluated using the MTT assay in the Caco-2 cell line. Finally, we examined the effect of these treatments along with Larazotide Acetate on the expression of genes involved in cell-tight junctions, such as Occludin, Claudin 3, tight junction protein-1, and Zonulin in the Caco-2 cell line. Our study demonstrated bromelain and ficin digestion effects on the commercial and wheat-extracted gliadin by SDS-PAGE, HPLC, and CD. Also, the cytotoxicity results on Caco-2 showed that toxicity of the gliadin and synthetic 19-mer peptide was decreased by adding bromelain and ficin. Furthermore, the proteolytic effects of bromelain and ficin on gliadin indicated the expression of genes involved in cell-tight junctions was improved. This study confirms that bromelain and ficin mixture could be effective in improving the symptoms of CeD.
Collapse
Affiliation(s)
- Masoumeh Sadat Mousavi Maleki
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Behrooz Vaziri
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Akbar Shabani
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ghazaleh Ghavami
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, 13198, Tehran, Iran
| | - Hamid Madanchi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, 13198, Tehran, Iran.
| | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, 13198, Tehran, Iran.
| |
Collapse
|
22
|
Clemente-Suárez VJ, Martín-Rodríguez A, Redondo-Flórez L, Villanueva-Tobaldo CV, Yáñez-Sepúlveda R, Tornero-Aguilera JF. Epithelial Transport in Disease: An Overview of Pathophysiology and Treatment. Cells 2023; 12:2455. [PMID: 37887299 PMCID: PMC10605148 DOI: 10.3390/cells12202455] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Epithelial transport is a multifaceted process crucial for maintaining normal physiological functions in the human body. This comprehensive review delves into the pathophysiological mechanisms underlying epithelial transport and its significance in disease pathogenesis. Beginning with an introduction to epithelial transport, it covers various forms, including ion, water, and nutrient transfer, followed by an exploration of the processes governing ion transport and hormonal regulation. The review then addresses genetic disorders, like cystic fibrosis and Bartter syndrome, that affect epithelial transport. Furthermore, it investigates the involvement of epithelial transport in the pathophysiology of conditions such as diarrhea, hypertension, and edema. Finally, the review analyzes the impact of renal disease on epithelial transport and highlights the potential for future research to uncover novel therapeutic interventions for conditions like cystic fibrosis, hypertension, and renal failure.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain;
- Group de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | | | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain; (L.R.-F.); (C.V.V.-T.)
| | - Carlota Valeria Villanueva-Tobaldo
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain; (L.R.-F.); (C.V.V.-T.)
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile;
| | | |
Collapse
|
23
|
Soheilian Khorzoghi M, Rostami-Nejad M, Yadegar A, Dabiri H, Hadadi A, Rodrigo L. Impact of probiotics on gut microbiota composition and clinical symptoms of coeliac disease patients following gluten-free diet. Contemp Clin Trials Commun 2023; 35:101201. [PMID: 37680267 PMCID: PMC10480319 DOI: 10.1016/j.conctc.2023.101201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/03/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Coeliac disease (CD) is associated with alterations in gut microbiota composition. This study evaluated the effects of probiotics on gut microbiota composition and clinical symptoms of treated CD patients. In this double-blind, placebo-controlled trial study, 31 CD patients that were randomly classified as probiotics (n = 15) and placebo (n = 16) groups received 109 colony-forming units/capsule for 12 weeks. Fecal samples were collected before and after probiotics, or placebo administration and the changes in intestinal microbiota were assessed by quantitative real-time PCR. Probiotic administration improved the patients' clinical symptoms when compared to the placebo group. Fatigue score was significantly reduced by the intake of probiotic supplements (P = 0.02). Except for Staphylococcus spp., the relative abundances of Bacteroidetes, Lactobacillus spp., Bifidobacterium spp., Clostridium cluster I, Enterobacteriaceae, and Firmicutes were higher in probiotics group. Accordingly, a 12-week multi-strain probiotic treatment regimen may modify the composition of intestinal microbiota and improve GI symptoms in CD patients.
Collapse
Affiliation(s)
| | - Mohammad Rostami-Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Dabiri
- Department of Microbiology, Faculty of Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Hadadi
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Luis Rodrigo
- Gastroenterology and Liver Service, Hospital Universitario Central de Asturias, School of Medicine, University of Oviedo, Spain
| |
Collapse
|
24
|
Vanoli A, Guerini C, Arpa G, Klersy C, Grillo F, Casadei Gardini A, De Hertogh G, Ferrante M, Moens A, Furlan D, Sessa F, Quaquarini E, Lenti MV, Neri G, Macciomei MC, Fassan M, Cascinu S, Paulli M, Graham RP, Di Sabatino A. Mismatch repair deficiency as prognostic factor for stage III small bowel adenocarcinoma: A multicentric international study. Dig Liver Dis 2023; 55:1261-1269. [PMID: 37236851 DOI: 10.1016/j.dld.2023.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Small bowel adenocarcinoma (SBA) is a rare cancer with an aggressive behavior. No study has specifically addressed the putative prognostic role of mismatch repair status in stage III SBAs. AIMS We aimed to investigate whether mismatch repair deficiency is associated with cancer-specific survival in a Western cohort of patients with stage III SBAs. METHODS In this retrospective multicentric international cohort study, we enrolled 70 patients who underwent surgically resection for stage III SBAs and we analyzed the frequency of mismatch repair deficiency, tested by immunohistochemistry for mismatch repair proteins and by polymerase chain reaction for microsatellite instability, and its association with cancer-specific survival and other clinic-pathologic factors. RESULTS We found sixteen (23%) patients with mismatch repair deficient adenocarcinoma, without discordance between immunohistochemical and polymerase chain reaction for microsatellite instability analyses. Mismatch repair deficiency proved to be associated with a better outcome both at univariable analysis (hazard ratio: 0.28, 95% confidence interval: 0.08-0.91, p: 0.035) and in bivariable models adjusted for patient age or gender, tumor site, pT4 stage, tumor budding, and perineural invasion. CONCLUSION This study highlights the importance of testing mismatch repair status to improve prognostic stratification in stage III SBAs.
Collapse
Affiliation(s)
- Alessandro Vanoli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy; Unit of Anatomic Pathology, Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | - Camilla Guerini
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | - Giovanni Arpa
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | - Catherine Klersy
- Clinical Epidemiology and Biometry, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia 27100, Italy
| | - Federica Grillo
- Pathology Unit, Department of Surgical and Diagnostic Sciences, University of Genoa and Ospedale Policlinico San Martino University Hospital, Genoa 16132, Italy
| | - Andrea Casadei Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan 20132, Italy
| | - Gert De Hertogh
- Department of Pathology, KU Leuven University Hospitals, Leuven 3000, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals, KU Leuven, Leuven 3000, Belgium
| | - Annick Moens
- Department of Gastroenterology and Hepatology, University Hospitals, KU Leuven, Leuven 3000, Belgium
| | - Daniela Furlan
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Fausto Sessa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Erica Quaquarini
- Medical Oncology Unit, ICS Maugeri-IRCCS SpA SB, Pavia 27100, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine, University of Pavia, San Matteo Hospital Foundation, Pavia 27100, Italy
| | - Giuseppe Neri
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | | | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine, DIMED, University of Padua, Padua 35122, Italy; Veneto Institute of Oncology, IOV-IRCCS, Padua 35128, Italy
| | - Stefano Cascinu
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan 20132, Italy
| | - Marco Paulli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy; Unit of Anatomic Pathology, Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | | | - Antonio Di Sabatino
- Department of Internal Medicine, University of Pavia, San Matteo Hospital Foundation, Pavia 27100, Italy.
| |
Collapse
|
25
|
Manai F, Zanoletti L, Arfini D, Micco SGD, Gjyzeli A, Comincini S, Amadio M. Dimethyl Fumarate and Intestine: From Main Suspect to Potential Ally against Gut Disorders. Int J Mol Sci 2023; 24:9912. [PMID: 37373057 DOI: 10.3390/ijms24129912] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Dimethyl fumarate (DMF) is a well-characterized molecule that exhibits immuno-modulatory, anti-inflammatory, and antioxidant properties and that is currently approved for the treatment of psoriasis and multiple sclerosis. Due to its Nrf2-dependent and independent mechanisms of action, DMF has a therapeutic potential much broader than expected. In this comprehensive review, we discuss the state-of-the-art and future perspectives regarding the potential repurposing of DMF in the context of chronic inflammatory diseases of the intestine, such as inflammatory bowel disorders (i.e., Crohn's disease and ulcerative colitis) and celiac disease. DMF's mechanisms of action, as well as an exhaustive analysis of the in vitro/in vivo evidence of its beneficial effects on the intestine and the gut microbiota, together with observational studies on multiple sclerosis patients, are here reported. Based on the collected evidence, we highlight the new potential applications of this molecule in the context of inflammatory and immune-mediated intestinal diseases.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Davide Arfini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Simone Giorgio De Micco
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Arolda Gjyzeli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
26
|
Lenti MV, Aronico N, Bianchi PI, D'Agate CC, Neri M, Volta U, Mumolo MG, Astegiano M, Calabrò AS, Zingone F, Latella G, Di Sario A, Carroccio A, Ciacci C, Luzza F, Bagnato C, Fantini MC, Elli L, Cammarota G, Gasbarrini A, Portincasa P, Latorre MA, Petrucci C, Quatraccioni C, Iannelli C, Vecchione N, Rossi CM, Broglio G, Ianiro G, Marsilio I, Bibbò S, Marinoni B, Tomaselli D, Abenavoli L, Pilia R, Santacroce G, Lynch E, Carrieri A, Mansueto P, Gabba M, Alunno G, Rossi C, Onnis F, Efthymakis K, Cesaro N, Vernero M, Baiano Svizzero F, Semeraro FP, Silano M, Vanoli A, Klersy C, Corazza GR, Di Sabatino A. Diagnostic delay in adult coeliac disease: An Italian multicentre study. Dig Liver Dis 2023; 55:743-750. [PMID: 36567177 DOI: 10.1016/j.dld.2022.11.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND There are few data regarding the diagnostic delay and its predisposing factors in coeliac disease (CD). AIMS To investigate the overall, the patient-dependant, and the physician-dependant diagnostic delays in CD. METHODS CD adult patients were retrospectively enroled at 19 Italian CD outpatient clinics (2011-2021). Overall, patient-dependant, and physician-dependant diagnostic delays were assessed. Extreme diagnostic, i.e., lying above the third quartile of our population, was also analysed. Multivariable regression models for factors affecting the delay were fitted. RESULTS Overall, 2362 CD patients (median age at diagnosis 38 years, IQR 27-46; M:F ratio=1:3) were included. The median overall diagnostic delay was 8 months (IQR 5-14), while patient- and physician-dependant delays were 3 (IQR 2-6) and 4 (IQR 2-6) months, respectively. Previous misdiagnosis was associated with greater physician-dependant (1.076, p = 0.005) and overall (0.659, p = 0.001) diagnostic delays. Neurological symptoms (odds ratio 2.311, p = 0.005) and a previous misdiagnosis (coefficient 9.807, p = 0.000) were associated with a greater extreme physician-dependant delay. Gastrointestinal symptoms (OR 1.880, p = 0.004), neurological symptoms (OR 2.313, p = 0.042), and previous misdiagnosis (OR 4.265, p = 0.000) were associated with increased extreme overall diagnostic delay. CONCLUSION We identified some factors that hamper CD diagnosis. A proper screening strategy for CD should be implemented.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Nicola Aronico
- First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Paola Ilaria Bianchi
- First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Carmela Cinzia D'Agate
- Department of Gastroenterology and Endoscopy, University Hospital "G. Rodolico", Catania, Italy
| | - Matteo Neri
- Department of Medicine and Ageing Sciences, 'G. d'Annunzio' University of Chieti-Pescara, Chieti, Italy
| | - Umberto Volta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Gloria Mumolo
- Department of General Surgery and Gastroenterology, Pisa University Hospital, Pisa, Italy
| | - Marco Astegiano
- SC Gastroenterologia AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Antonino Salvatore Calabrò
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" University of Florence, Florence, Italy
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, and Gastroenterology Unit, Azienda Ospedale Università, Padova, Italy
| | - Giovanni Latella
- Gastroenterology, Hepatology and Nutrition Division, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Di Sario
- Department of Gastroenterology and Transplantation, Polytechnic Marche University, Ancona, Italy
| | - Antonio Carroccio
- Internal Medicine Unit, "V. Cervello Hospital", University of Palermo, Palermo, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, Gastroenterology Unit, University of Salerno, Salerno, Italy
| | - Francesco Luzza
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Carmela Bagnato
- Clinical Nutrition Unit, Madonna delle Grazie Hospital, Matera, Italy
| | | | - Luca Elli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giovanni Cammarota
- Digestive Disease Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Medicina Interna e Gastroenterologia, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Medicina Interna e Gastroenterologia, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Clinica Medica "A. Murri", Bari, Italy
| | - Mario Andrea Latorre
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Clarissa Petrucci
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Claudia Quatraccioni
- Gastroenterology, Hepatology and Nutrition Division, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Chiara Iannelli
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Nicoletta Vecchione
- Department of Gastroenterology and Endoscopy, University Hospital "G. Rodolico", Catania, Italy
| | - Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giacomo Broglio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gianluca Ianiro
- Digestive Disease Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Medicina Interna e Gastroenterologia, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Ilaria Marsilio
- Department of Surgery, Oncology and Gastroenterology, University of Padua, and Gastroenterology Unit, Azienda Ospedale Università, Padova, Italy
| | - Stefano Bibbò
- Digestive Disease Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Medicina Interna e Gastroenterologia, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Beatrice Marinoni
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Riccardo Pilia
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
| | - Giovanni Santacroce
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Erica Lynch
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" University of Florence, Florence, Italy
| | - Antonella Carrieri
- Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Clinica Medica "A. Murri", Bari, Italy
| | - Pasquale Mansueto
- Internal Medicine Unit, "V. Cervello Hospital", University of Palermo, Palermo, Italy
| | - Margherita Gabba
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giacomo Alunno
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Rossi
- Department of Gastroenterology and Transplantation, Polytechnic Marche University, Ancona, Italy
| | - Francesca Onnis
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Konstantinos Efthymakis
- Department of Medicine and Ageing Sciences, 'G. d'Annunzio' University of Chieti-Pescara, Chieti, Italy
| | - Nicola Cesaro
- Gastroenterology, Hepatology and Nutrition Division, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marta Vernero
- SC Gastroenterologia AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | | | | | | | - Alessandro Vanoli
- Unit of Anatomic Pathology, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Catherine Klersy
- Clinical Epidemiology and Biometry Service, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
27
|
Maurano F, Rotondi Aufiero V, Treppiccione L, Rossi S, Luongo D, Mazzarella G, Rossi M. The HLA-DQ8 transgenic mouse: A model to study the immune and cytotoxic responses to wheat gliadin. Methods Cell Biol 2023; 179:157-171. [PMID: 37625873 DOI: 10.1016/bs.mcb.2023.01.007] [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] [Indexed: 08/27/2023]
Abstract
A complete understanding of celiac disease (CD) pathogenesis has been hindered to date because of the lack of adequate in vivo models. Herein, we describe two in vivo approaches in HLA-DQ8-transgenic mice to study the intrinsic cytoxicity and immune features of wheat gliadin. By adopting the first method, we explored the mucosal architecture of the small intestine following the intra-gastric administration of wheat gliadin in mice treated with indomethacin, an inhibitor of cyclooxygenases. Mice showed a significant reduction of villus height, increased crypt depth and increased intraepithelial lymphocytes. The second approach involved the mucosal sensitization to gliadin via the intranasal route. This protocol induced a Th1/Th17 phenotype in mesenteric lymph nodes, as described in CD. In conclusion, these methods remain instrumental to analyze in vivo distinct biological features of wheat gliadin and related prolamins. Furthermore, the sensitization protocol could be exploited to test innovative strategies downregulating the gliadin-specific immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
28
|
Mulder CJJ, Elli L, Lebwohl B, Makharia GK, Rostami K, Rubio-Tapia A, Schumann M, Tye-Din J, Zeitz J, Al-Toma A. Follow-Up of Celiac Disease in Adults: "When, What, Who, and Where". Nutrients 2023; 15:2048. [PMID: 37432208 PMCID: PMC10181343 DOI: 10.3390/nu15092048] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
For patients with celiac disease (CeD), a lifelong gluten-free diet is not a voluntary lifestyle choice-it is a necessity. The key end points in clinical follow-up are symptom resolution, the normalization of weight, prevention of overweight, seroconversion, and negation or minimization of increased long-term morbidity. For the latter, a surrogate endpoint is mucosal healing, which means the normalization of histology to Marsh 0-1. Ideally, celiac follow-up care includes a multidisciplinary approach, effective referral processes, improved access that leverages technological advances, and following guidelines with the identification of measurable quality indicators, ideally informed by evidence-based research. Face-to-face CeD care and telemedicine are considered the standards for this process, although published data are insufficient. Guidelines and statements on diagnosis are readily available. However, data are lacking on optimal clinic visit intervals and outcomes and quality indicators such as improvement of symptoms, function and quality of life, survival and disease control, and how to most effectively use healthcare resources. The results of future research should provide the basis for general recommendations for evidence-based standards of quality of care in CeD.
Collapse
Affiliation(s)
- Chris J. J. Mulder
- Department of Gastroenterology, Amsterdam UMC, Location Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Benjamin Lebwohl
- Department of Medicine, Celiac Disease Center, Columbia University Medical Center, 180 Fort Washington Avenue, Suite 936, New York, NY 10032, USA;
| | - Govind K. Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India;
| | - Kamran Rostami
- Department of Gastroenterology, Palmerston North Hospital, Palmerston North 4442, New Zealand;
| | - Alberto Rubio-Tapia
- Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany;
| | - Jason Tye-Din
- Department of Immunology, The Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia;
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Jonas Zeitz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
- Swiss Celiac Center, Center of Gastroenterology, Clinic Hirslanden, 8032 Zurich, Switzerland
| | - Abdulbaqi Al-Toma
- Department of Gastroenterology and Hepatology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands;
| |
Collapse
|
29
|
Tedaldi G, Guerini C, Angeli D, Furlan D, Libera L, Lenti MV, Grillo F, Fassan M, Solcia E, Sessa F, Paulli M, Di Sabatino A, Ulivi P, Vanoli A. Molecular Landscape and Association With Crohn Disease of Poorly Cohesive Carcinomas of the Nonampullary Small Bowel. Am J Clin Pathol 2023; 159:315-324. [PMID: 36812376 PMCID: PMC10071142 DOI: 10.1093/ajcp/aqac161] [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: 08/18/2022] [Accepted: 12/14/2022] [Indexed: 02/24/2023] Open
Abstract
OBJECTIVES Poorly cohesive carcinomas (PCCs) are neoplasms defined by a predominantly dyshesive growth pattern with single cell or cord-like stromal infiltration. The -distinctive clinicopathologic and prognostic features of small bowel PCCs (SB-PCCs) in comparison with conventional-type small intestinal adenocarcinomas have only recently been characterized. However, as SB-PCCs' genetic profile is still unknown, we aimed to analyze the molecular landscape of SB-PCCs. METHODS A next-generation sequencing analysis through Trusight Oncology 500 on a series of 15 nonampullary SB-PCCs was performed. RESULTS The most frequently found gene alterations were TP53 (53%) and RHOA (13%) mutations and KRAS amplification (13%), whereas KRAS, BRAF, and PIK3CA mutations were not identified. Most SB-PCCs (80%) were associated with Crohn disease, including both RHOA-mutated SB-PCCs, which featured a non-SRC-type histology, and showed a peculiar appendiceal-type, low-grade goblet cell adenocarcinoma (GCA)-like component. Rarely, SB-PCCs showed high microsatellite instability, mutations in IDH1 and ERBB2 genes, or FGFR2 amplification (one case each), which are established or promising therapeutic targets in such aggressive cancers. CONCLUSIONS SB-PCCs may harbor RHOA mutations, which are reminiscent of the diffuse subtype of gastric cancers or appendiceal GCAs, while KRAS and PIK3CA mutations, commonly involved in colorectal and small bowel adenocarcinomas, are not typical of such cancers.
Collapse
Affiliation(s)
- Gianluca Tedaldi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”Meldola, Italy
| | - Camilla Guerini
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”Meldola, Italy
| | - Daniela Furlan
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Laura Libera
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Federica Grillo
- Department of Laboratory Services, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Surgical and Diagnostic Sciences (DISC), Pathology Unit, University of Genova, Genova, Italy
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padua, Italy
- Veneto Institute of Oncology (I.O.V. IRCCS), Padua, Italy
| | - Enrico Solcia
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
| | - Fausto Sessa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Marco Paulli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”Meldola, Italy
| | - Alessandro Vanoli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
30
|
Manai F, Zanoletti L, Morra G, Mansoor S, Carriero F, Bozzola E, Muscianisi S, Comincini S. Gluten Exorphins Promote Cell Proliferation through the Activation of Mitogenic and Pro-Survival Pathways. Int J Mol Sci 2023; 24:3912. [PMID: 36835317 PMCID: PMC9966116 DOI: 10.3390/ijms24043912] [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: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Celiac disease (CD) is a chronic and systemic autoimmune disorder that affects preferentially the small intestine of individuals with a genetic predisposition. CD is promoted by the ingestion of gluten, a storage protein contained in the endosperm of the seeds of wheat, barley, rye, and related cereals. Once in the gastrointestinal (GI) tract, gluten is enzymatically digested with the consequent release of immunomodulatory and cytotoxic peptides, i.e., 33mer and p31-43. In the late 1970s a new group of biologically active peptides, called gluten exorphins (GEs), was discovered and characterized. In particular, these short peptides showed a morphine-like activity and high affinity for the δ-opioid receptor (DOR). The relevance of GEs in the pathogenesis of CD is still unknown. Recently, it has been proposed that GEs could contribute to asymptomatic CD, which is characterized by the absence of symptoms that are typical of this disorder. In the present work, GEs cellular and molecular effects were in vitro investigated in SUP-T1 and Caco-2 cells, also comparing viability effects with human normal primary lymphocytes. As a result, GEs treatments increased tumor cell proliferation by cell cycle and Cyclins activation as well as by induction of mitogenic and pro-survival pathways. Finally, a computational model of GEs interaction with DOR is provided. Altogether, the results might suggest a possible role of GEs in CD pathogenesis and on its associated cancer comorbidities.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
- Laboratory for Mucosal Immunology, TARGID, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Morra
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Samman Mansoor
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Elena Bozzola
- Pediatric Unit, I.R.C.C.S. Bambino Gesù Children Hospital, 00165 Roma, Italy
| | - Stella Muscianisi
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
31
|
The Effect of a Gluten-Free Diet on Vitamin D Metabolism in Celiac Disease: The State of the Art. Metabolites 2023; 13:metabo13010074. [PMID: 36676999 PMCID: PMC9861273 DOI: 10.3390/metabo13010074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Celiac disease is a chronic autoimmune disorder involving the small intestine, characterized by villous atrophy, crypt hyperplasia and an increase in intraepithelial lymphocytes. Due to both calcium malabsorption and immune activation, a high prevalence of bone mass derangement is evident in this condition, regardless of the presence of overt malabsorption. Alterations of mineral metabolism are also frequently described, and in this review, the modifications of serum levels of vitamin D are analyzed, according to the available literature on this topic. In untreated patients, secondary hyperparathyroidism is responsible for the hyperconversion of 25-vitamin D into 1,25-vitamin D making mandatory the determination of serum levels of both vitamin metabolites to avoid a wrong diagnosis of vitamin D deficit. A gluten-free diet allows for a normalization of bone and mineral metabolism, reverting these abnormalities and raising some doubts on the need for vitamin supplementation in all the patients. Data available do not support this wide indication, and a complete evaluation of bone and mineral metabolism should be performed to select patients who need this therapeutic approach.
Collapse
|
32
|
Rubio-Tapia A, Hill ID, Semrad C, Kelly CP, Greer KB, Limketkai BN, Lebwohl B. American College of Gastroenterology Guidelines Update: Diagnosis and Management of Celiac Disease. Am J Gastroenterol 2023; 118:59-76. [PMID: 36602836 DOI: 10.14309/ajg.0000000000002075] [Citation(s) in RCA: 167] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/23/2022] [Indexed: 01/06/2023]
Abstract
This guideline presents an update to the 2013 American College of Gastroenterology Guideline on the Diagnosis and Management of Celiac Disease with updated recommendations for the evaluation and management of patients with celiac disease (CD). CD is defined as a permanent immune-mediated response to gluten present in wheat, barley, and rye. CD has a wide spectrum of clinical manifestations that resemble a multisystemic disorder rather than an isolated intestinal disease, and is characterized by small bowel injury and the presence of specific antibodies. Detection of CD-specific antibodies (e.g., tissue transglutaminase) in the serum is very helpful for the initial screening of patients with suspicion of CD. Intestinal biopsy is required in most patients to confirm the diagnosis. A nonbiopsy strategy for the diagnosis of CD in selected children is suggested and discussed in detail. Current treatment for CD requires strict adherence to a gluten-free diet (GFD) and lifelong medical follow-up. Most patients have excellent clinical response to a GFD. Nonresponsive CD is defined by persistent or recurrent symptoms despite being on a GFD. These patients require a systematic workup to rule out specific conditions that may cause persistent or recurrent symptoms, especially unintentional gluten contamination. Refractory CD is a rare cause of nonresponsive CD often associated with poor prognosis.
Collapse
Affiliation(s)
- Alberto Rubio-Tapia
- Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ivor D Hill
- Division of Gastroenterology, Hepatology, and Nutrition, Nationwide Children Hospital, Columbus, Ohio, USA
| | - Carol Semrad
- Division of Gastroenterology, University of Chicago, Chicago, Illinois, USA
| | - Ciarán P Kelly
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Katarina B Greer
- Department of Medicine, Section of Gastroenterology and Hepatology, Louis Stokes VA Medical Center, Cleveland, Ohio, USA
| | - Berkeley N Limketkai
- Division of Digestive Diseases, UCLA School of Medicine, Los Angeles, California, USA
| | - Benjamin Lebwohl
- Division of Gastroenterology and Hepatology, Columbia University, New York, USA
| |
Collapse
|
33
|
Lal R, Bhardwaj R, Minz RW, Prasad KK, Lal S, Dayal D, Kumar Y. Usefulness of a double immunofluorescence technique for detection of intestinal tTG-IgA deposits in diabetic and non-diabetic children with celiac disease. Pediatr Neonatol 2022:S1875-9572(22)00269-8. [PMID: 36610914 DOI: 10.1016/j.pedneo.2022.01.012] [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: 09/18/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Celiac disease (CD) is frequently associated with type I diabetes mellitus (T1D), where its diagnosis may be a challenging task. This study aims to test the usefulness of the double staining immunofluorescence (dsIF) technique for the detection of intestinal anti-tissue transglutaminase specific IgA antibody (tTG-IgA) deposits in CD and T1D children with coexisting CD. METHODS A total of 46 patients (30 cases of CD and 16 cases of T1D with CD) and 16 non-diabetic, non-celiac children were recruited. Endoscopic biopsies were taken and analyzed by light microscopy, quantitative histology (QH), and a dsIF technique. RESULTS Histologically, villous atrophy was most severe in CD, followed by T1D with CD, while all control biopsies except 1 were normal. QH showed a statistically significant difference in villous height (Vh), crypt depth (CrD), and Vh:CrD ratio between diabetic and non-diabetic patients with CD. dsIF technique could detect tTG-IgA deposits in 85.7% of cases of CD alone and 93.8% of biopsies from diabetic children. Surprisingly, deposits were more extensive in biopsies with minimal villous shortening. Also, all 5 biopsies from T1D patients with normal histology were dsIF positive. CONCLUSION In-situ analysis of tTG-IgA immune deposits facilitates the detection of positive serology early-onset CD. Quantitative analysis may be used as an ancillary tool to increase the reliability of histological findings in these patients.
Collapse
Affiliation(s)
- Raghav Lal
- Department of Immunopathology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Ranjeet Bhardwaj
- Department of Immunopathology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Ranjana Walker Minz
- Department of Immunopathology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Kaushal Kishore Prasad
- Department of Gastroenterology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Sadhna Lal
- Department of Pediatric Gastroenterology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Devi Dayal
- Department of Pediatric Endocrinology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Yashwant Kumar
- Department of Immunopathology, Post Graduate Institute of Medical Education & Research, Chandigarh, India.
| |
Collapse
|
34
|
Maddah R, Panji A, Khoei MA, Bazireh H, Abedinlou H, Davodabadi F, Shariati P. Deciphering potential biomarkers for celiac disease by using an integrated bioinformatics approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
35
|
Lenti MV, Rossi CM, Melazzini F, Gastaldi M, Bugatti S, Rotondi M, Bianchi PI, Gentile A, Chiovato L, Montecucco C, Corazza GR, Di Sabatino A. Seronegative autoimmune diseases: A challenging diagnosis. Autoimmun Rev 2022; 21:103143. [PMID: 35840037 DOI: 10.1016/j.autrev.2022.103143] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/10/2022] [Indexed: 12/19/2022]
Abstract
Autoimmune diseases (AID) are increasingly prevalent conditions which comprise more than 100 distinct clinical entities that are responsible for a great disease burden worldwide. The early recognition of these diseases is key for preventing their complications and for tailoring proper management. In most cases, autoantibodies, regardless of their potential pathogenetic role, can be detected in the serum of patients with AID, helping clinicians in making a definitive diagnosis and allowing screening strategies for early -and sometimes pre-clinical- diagnosis. Despite their undoubted crucial role, in a minority of cases, patients with AID may not show any autoantibody, a condition that is referred to as seronegative AID. Suboptimal accuracy of the available laboratory tests, antibody absorption, immunosuppressive therapy, immunodeficiencies, antigen exhaustion, and immunosenescence are the main possible determinants of seronegative AID. Indeed, in seronegative AID, the diagnosis is more challenging and must rely on clinical features and on other available tests, often including histopathological evaluation and radiological diagnostic tests. In this review, we critically dissect, in a narrative fashion, the possible causes of seronegativity, as well as the diagnostic and management implications, in several AID including autoimmune gastritis, celiac disease, autoimmune liver disease, rheumatoid arthritis, autoimmune encephalitis, myasthenia gravis, Sjögren's syndrome, antiphospholipid syndrome, and autoimmune thyroid diseases.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Federica Melazzini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | - Serena Bugatti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Unit of Rheumatology, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Mario Rotondi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
| | - Paola Ilaria Bianchi
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Antonella Gentile
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Luca Chiovato
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Unit of Rheumatology, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy.
| |
Collapse
|
36
|
Catassi C, Verdu EF, Bai JC, Lionetti E. Coeliac disease. Lancet 2022; 399:2413-2426. [PMID: 35691302 DOI: 10.1016/s0140-6736(22)00794-2] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 12/14/2022]
Abstract
Coeliac disease is an autoimmune disorder that primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. Prevalence in the general population ranges from 0·5% to 2%, with an average of about 1%. The development of the coeliac enteropathy depends on a complex immune response to gluten proteins, including both adaptive and innate mechanisms. Clinical presentation of coeliac disease is highly variable and includes classical and non-classical gastrointestinal symptoms, extraintestinal manifestations, and subclinical cases. The disease is associated with a risk of complications, such as osteoporosis and intestinal lymphoma. Diagnosis of coeliac disease requires a positive serology (IgA anti-transglutaminase 2 and anti-endomysial antibodies) and villous atrophy on small-intestinal biopsy. Treatment involves a gluten-free diet; however, owing to the high psychosocial burden of such a diet, research into alternative pharmacological treatments is currently very active.
Collapse
Affiliation(s)
- Carlo Catassi
- Department of Specialized Clinical Sciences and Odontostomatology, Polytechnic University of Marche, Ancona, Italy; Celiac Center and Mucosal Immunology and Biology Research, MassGeneral Hospital for Children-Harvard Medical School, Boston, MA, USA.
| | - Elena F Verdu
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Julio Cesar Bai
- Department of Medicine, Dr C Bonorino Udaondo Gastroenterology Hospital, Buenos Aires, Argentina; Research Institutes, Universidad del Salvador, Buenos Aires, Argentina
| | - Elena Lionetti
- Department of Specialized Clinical Sciences and Odontostomatology, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
37
|
Rasmussen SN, Vigre H, Madsen CB. Risk of consuming products with or without precautionary wheat or gluten labelling for persons with coeliac disease. Food Chem Toxicol 2022; 166:113231. [PMID: 35710030 DOI: 10.1016/j.fct.2022.113231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 10/18/2022]
Abstract
Persons with Coeliac disease (CD) need to keep a lifelong diet without gluten and therefore need to know which foods are safe to eat. The objective of this study was to analyse ordinary foods products without gluten containing ingredients (PWG) with or without the precautionary allergen statement (PAL) "May contain wheat/gluten" or similar wording and use these data to perform a probabilistic risk assessment. Foods with and without PAL were analysed for gluten. Inputs for the risk assessment were analytical results, daily food consumption and data from a 90-days challenge study in coeliac patients (Catassi et al., 2007).20/77 products with PAL and 10/51 without PAL had gluten ≥5 mg/kg 3/77 with PAL and 3/51 without PAL had gluten ≥20 mg/kg. A coconut cookie brand was outlier with 421 mg gluten/kg. The likelihood of developing mucosal damage in the CD population when eating the same PWG for 90 days in addition to certified gluten-free products was low (0.2-3.2%) with little difference between with or without PAL. Including the coconut cookie results increased the risk to 3.8-9.2%. Caution should be taken in connection to foods where there is an obvious chance of contamination.
Collapse
Affiliation(s)
- Sisse Nygaard Rasmussen
- National Food Institute, Technical University of Denmark, Denmark; New Nordic Manufacturing ApS, Denmark
| | - Håkan Vigre
- National Food Institute, Technical University of Denmark, Denmark
| | | |
Collapse
|
38
|
Follow-Up of a Rare Case of Eosinophilic Gastroenteritis Associated with Persistent Blood Eosinophilia and Multiple Food Allergies. Diagnostics (Basel) 2022; 12:diagnostics12061381. [PMID: 35741191 PMCID: PMC9221940 DOI: 10.3390/diagnostics12061381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Eosinophilic gastroenteritis (EGE) is a subgroup of the eosinophilic gastro-intestinal disorders (EGIDs), characterized by eosinophilic infiltration and chronic inflammation of the gastrointestinal tract. These are rare diseases with still incompletely elucidated causes and mechanisms, with frequently delayed diagnosis and variable outcome. Despite increased interest in eosinophilic diseases in recent years, fewer data have been published on EGE and no standardized diagnostic and therapeutic approach exists. This paper reports the case of a young male patient diagnosed with EGE in 2017 based on clinical and histopathological criteria and constantly monitored during five years. Besides gastrointestinal eosinophilic infiltration, biopsies also revealed eosinophilic infiltration of the oesophagus, despite no declared characteristic oesophageal symptoms. We found increased specific IgE to multiple foods and progressive blood hypereosinophilia which preceded EGE diagnosis by three years. The EGE management included selective dietary restrictions and pharmacologic therapy based on daily budesonide non-enteric coated tablets, proton pumps inhibitors, antihistamines, cromoglycate, correction of iron, calcium and vitamin D deficiencies. The clinical outcome was good, while blood eosinophilia and endoscopic appearance remained almost unchanged. After one year the patient complained of respiratory symptoms suggesting asthma, needing continuous combined inhaled therapy. The reported case is illustrative for complex presentation, diagnosis and outcome of a rare case of mucosal chronic EGE associated with oesophageal involvement, peripheral eosinophilia, multiple food allergies and asthma.
Collapse
|
39
|
Liu J, Lundemann AKJ, Reibel J, Pedersen AML. Salivary gland involvement and oral health in patients with coeliac disease. Eur J Oral Sci 2022; 130:e12861. [PMID: 35247226 PMCID: PMC9314853 DOI: 10.1111/eos.12861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/15/2022] [Indexed: 01/25/2023]
Abstract
Coeliac disease (CD) is a chronic immune‐mediated enteropathy triggered by ingestion of gluten. The aim of this study was to investigate if the salivary glands as a component of the mucosal immune system are involved in CD, leading to sialadenitis and salivary gland dysfunction and associated oral manifestations. Twenty patients with CD aged 49.2 (SD 15.5 years) and 20 age‐ and gender‐matched healthy controls underwent an interview regarding general and oral health, serological analysis, a clinical oral examination including bitewing radiographs, Candida smear, assessment of salivary mutans streptococci and lactobacilli levels, unstimulated and chewing‐stimulated whole and parotid saliva flow rates, analysis of secretory IgA, and a labial salivary gland biopsy. Xerostomia, mucosal lesions, dry/cracked lips and focal lymphocytic sialadenitis were more prevalent and extensive in patients with CD than in healthy controls. Moreover, the patients had less gingival inflammation and higher whole saliva flow rates than the healthy controls, but did not differ regarding dental health and levels of cariogenic bacteria and Candida. The major salivary gland function appears unaffected, contributing to maintenance of a balanced microbiota and oral health in CD patients. Xerostomia and labial dryness may be related to minor salivary gland inflammation and subsequent impaired mucosal lubrication.
Collapse
Affiliation(s)
- Jason Liu
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Kristine Juncker Lundemann
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Reibel
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Marie Lynge Pedersen
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Amnuaycheewa P, Abdelmoteleb M, Wise J, Bohle B, Ferreira F, Tetteh AO, Taylor SL, Goodman RE. Development of a Sequence Searchable Database of Celiac Disease-Associated Peptides and Proteins for Risk Assessment of Novel Food Proteins. FRONTIERS IN ALLERGY 2022; 3:900573. [PMID: 35769554 PMCID: PMC9234867 DOI: 10.3389/falgy.2022.900573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/15/2022] [Indexed: 12/29/2022] Open
Abstract
Celiac disease (CeD) is an autoimmune enteropathy induced by prolamin and glutelin proteins in wheat, barley, rye, and triticale recognized by genetically restricted major histocompatibility (MHC) receptors. Patients with CeD must avoid consuming these proteins. Regulators in Europe and the United States expect an evaluation of CeD risks from proteins in genetically modified (GM) crops or novel foods for wheat-related proteins. Our database includes evidence-based causative peptides and proteins and two amino acid sequence comparison tools for CeD risk assessment. Sequence entries are based on the review of published studies of specific gluten-reactive T cell activation or intestinal epithelial toxicity. The initial database in 2012 was updated in 2018 and 2022. The current database holds 1,041 causative peptides and 76 representative proteins. The FASTA sequence comparison of 76 representative CeD proteins provides an insurance for possible unreported epitopes. Validation was conducted using protein homologs from Pooideae and non-Pooideae monocots, dicots, and non-plant proteins. Criteria for minimum percent identity and maximum E-scores are guidelines. Exact matches to any of the 1,041 peptides suggest risks, while FASTA alignment to the 76 CeD proteins suggests possible risks. Matched proteins should be tested further by CeD-specific CD4/8+ T cell assays or in vivo challenges before their use in foods.
Collapse
Affiliation(s)
- Plaimein Amnuaycheewa
- Department of Agro-Industrial, Food, and Environmental Technology, King Mongkut's University of Technology North Bangkok (KMUTNB), Bangkok, Thailand
| | | | - John Wise
- Food Allergy Research and Resource Program (FARRP), Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Barbara Bohle
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | | - Steve L. Taylor
- Food Allergy Research and Resource Program (FARRP), Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Richard E. Goodman
- Food Allergy Research and Resource Program (FARRP), Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
- *Correspondence: Richard E. Goodman
| |
Collapse
|
41
|
Lenti MV, Vanoli A, Miceli E, Arpa G, Di Stefano M, Soriano S, Capuano F, Gentile A, Aronico N, Coppola L, Pasini A, Luinetti O, Mauro A, Paulli M, Klersy C, Corazza GR, Di Sabatino A. Increase of Deep Intraepithelial Lymphocytes in the Oxyntic Mucosa of Patients With Potential and Overt Autoimmune Gastritis. Front Immunol 2022; 13:866167. [PMID: 35603187 PMCID: PMC9114815 DOI: 10.3389/fimmu.2022.866167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/31/2022] [Indexed: 01/23/2023] Open
Abstract
Pathological correlates of potential autoimmune gastritis (AIG), defined by anti-parietal cell antibody (PCA) positivity in the absence of gastric atrophy, have never been described. We herein aimed to assess intraepithelial lymphocyte (IEL) infiltration in gastric corpus of AIG patients. From 2000 to 2021, among 53 potential AIG patients, we focused on nine (median age 61 years, IQR 53-82; four females) who subsequently developed overt AIG. IEL infiltration of the oxyntic mucosa was assessed before and after developing overt AIG by measuring deep and superficial CD3+ IEL. AIG patients with different degrees of corpus atrophy, healthy controls (HC), active H. pylori gastritis, celiac disease (CD), and Hashimoto’s thyroiditis patients were included as controls. Of note, deep, but not superficial, CD3+ IEL count was higher (p<0.001) in potential AIG compared to HC and H. pylori gastritis. Deep CD3+ IEL infiltration did not change before or after the evolution into atrophy (median 9.6, IQR 8.8-12.4, vs 11.3, IQR 9.4-12.9). No difference was found in deep CD3+ IEL infiltration among potential, mild, and severe AIG, and compared to Hashimoto’s thyroiditis or CD. A deep CD3+ IEL cut-off of >7/100 epithelial cells allowed discrimination of any AIG stage and severity (AUC=0.842). We conclude that an increased deep CD3+ IEL infiltration of the oxyntic mucosa could represent a marker of potential AIG. Prospective studies including a larger number of potential AIG patients are needed.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Emanuela Miceli
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Giovanni Arpa
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Michele Di Stefano
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Simone Soriano
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Francesca Capuano
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Antonella Gentile
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Nicola Aronico
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Luigi Coppola
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandra Pasini
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Ombretta Luinetti
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Aurelio Mauro
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marco Paulli
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Catherine Klersy
- Unit of Clinical Epidemiology and Biometry, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| |
Collapse
|
42
|
Saha BK, Saha S, Bonnier A, Saha BN. Association between idiopathic pulmonary hemosiderosis and celiac disease in pediatric patients: A scoping review of the literature over the past 50 years. Pediatr Pulmonol 2022; 57:1127-1144. [PMID: 35088581 DOI: 10.1002/ppul.25847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Idiopathic pulmonary hemosiderosis (IPH) is a rare cause of diffuse alveolar hemorrhage, the mechanism of which is currently unknown. Nearly one-third of pediatric patients with IPH test positive for Celiac disease (CD) serology. Several hypothetical mechanisms have been proposed to unify the coexistence of these two entities, also referred to as Lane-Hamilton syndrome (LHS). METHOD This manuscript is a scoping review of the medical literature. Medline, Embase, and PubMed Central databases were searched between 1971 and 2021 with appropriate search words to identify all cases of pediatric LHS. RESULTS A total of 20 manuscripts with 23 pediatric patients with LHS were identified. The mean age was 11 years, and 13/23 (56.5%) of the children were boys. Hemoptysis was present in 57% of patients during diagnosis. Bronchoscopy with bronchoalveolar lavage demonstrating hemosiderin laden macrophages was the primary mode of diagnostic confirmation. Only three patients underwent lung biopsy. Any significant GI symptom was reported in a minority of patients (22%). Iron deficiency anemia on presentation was described in 83% of children. The majority of patients were malnourished. Serology for CD was positive in all patients, as was the histopathologic analysis of the small bowel biopsy. No patients had any other autoantibody positivity. The introduction of gluten free diet (GFD) was associated with a positive response in 20/23 patients. CONCLUSION All pediatric patients with IPH should undergo screening for CD. Low serum ferritin in patients with IPH could be suggestive of coexisting CD. Strict GFD should be tried as the initial therapy.
Collapse
Affiliation(s)
- Biplab K Saha
- Department of Pulmonary and Critical Care Medicine, Ozarks Medical Center, West Plains, Missouri, USA
| | - Santu Saha
- Department of Internal Medicine, Bangladesh Medical College, Dhaka, Bangladesh
| | - Alyssa Bonnier
- Department of Critical Care Nursing, Goldfarb School of Nursing, Barnes Jewish College, St. Louis, Missouri, USA
| | | |
Collapse
|
43
|
Lenti MV, Miceli E, Vanoli A, Klersy C, Corazza GR, Di Sabatino A. Time course and risk factors of evolution from potential to overt autoimmune gastritis. Dig Liver Dis 2022; 54:642-644. [PMID: 34732311 DOI: 10.1016/j.dld.2021.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/26/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The natural history of patients with potential autoimmune gastritis (AIG), defined by the presence of serum anti-parietal cell antibody (PCA) positivity and no gastric histopathological alterations, is unknown. We therefore aimed to assess the natural history and clinical correlates of potential autoimmune gastritis (AIG). METHODS In 2000-2019, we enrolled potential AIG patients by monitoring once a year (±6 months) histopathological evolution into overt AIG, defined as the occurrence of atrophy in the oxyntic mucosa. Factors affecting disease progression were assessed. RESULTS Fifty-one potential AIG patients (median age 57 years, IQR 43-73, F:M ratio 1.7:1) were monitored for up to 15 years (median 6 years, IQR 3-8). Of them, 24 (47.1%) evolved into overt AIG in a median time of 2 years (IQR 2-4.5). Having a concomitant autoimmune disorder (HR 4.09, 95% CI 1.52-11.00; p = 0.005), but not older age (HR 1.00, 95% CI 0.45-2.22; p = 0.992) and female sex (HR 1.19, 95% CI 0.51-2.78; p = 0.395), was associated with evolution into overt AIG. CONCLUSIONS Roughly one in two potential AIG patients will evolve into overt AIG over a median time of two years, especially those with a concurrent autoimmune disorder.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Emanuela Miceli
- Department of Internal Medicine, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Department of Pathology, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Catherine Klersy
- Unit of Clinical Epidemiology and Biometry, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy.
| |
Collapse
|
44
|
Nazario E, Lasa J, Schill A, Duarte B, Berardi D, Paz S, Muryan A, Zubiaurre I. IgA Deficiency Is Not Systematically Ruled Out in Patients Undergoing Celiac Disease Testing. Dig Dis Sci 2022; 67:1238-1243. [PMID: 33770329 DOI: 10.1007/s10620-021-06939-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 03/06/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Guidelines for celiac disease (CD) testing recommend total serum IgA determination alongside anti-transglutaminase IgA antibodies. It is not well known if lack of serum IgA determination is a common finding in clinical practice. AIM To determine the prevalence of lack of serum IgA determination among patients screened for celiac disease. MATERIALS AND METHODS We identified all subjects who underwent serum anti-transglutaminase IgA and/or other CD-related antibodies determination at a single teaching hospital in Buenos Aires from October 2019 to February 2020. Medical records were reviewed to select adult patients who were tested for celiac disease. The primary outcome was the proportion of patients with inadequate testing for celiac disease due to lack of serum IgA determination. We retrieved the following variables from each patient's record: age, gender, body mass index, symptoms present at screening, first-grade family history of CD, history of type-1 diabetes mellitus, autoimmune hypothyroidism, Down's syndrome. RESULTS Overall, 1122 patients were included for analysis. Lack of serum IgA determination prevalence was 20.49%. Among patients who did have serum IgA determination, the prevalence of IgA deficiency was 5.16%. The following variables were independently associated with a significantly increased odds of serum IgA determination: diarrhea [OR 1.55 (1.01-2.34)] and abdominal pain [OR 2.28 (1.44-3.63)]; higher body mass index [OR 0.91 (0.85-0.98)], osteoporosis [OR 0.49 (0.28-0.89)], hypothyroidism [OR 0.18 (0.07-0.45)], arthralgia/arthritis [OR 0.47 (0.27-0.85)], or testing by endocrinologist [OR 0.46 (0.23-0.91)] and gynecologist [OR 0.14 (0.06-0.31)] were inversely associated. CONCLUSION IgA deficiency is not systematically ruled out in a relatively high proportion of patients undergoing serological screening of celiac disease.
Collapse
Affiliation(s)
- Ezequiel Nazario
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina
| | - Juan Lasa
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina.
| | - Amalia Schill
- Biochemistry Department, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Belen Duarte
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina
| | - Diego Berardi
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina
| | - Silvina Paz
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina
| | - Alexis Muryan
- Biochemistry Department, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Ignacio Zubiaurre
- Gastroenterology Department, Hospital Británico de Buenos Aires, Perdriel 74 (1012), Buenos Aires, Argentina
| |
Collapse
|
45
|
Vanoli A, Guerini C, Grillo F, Klersy C, Fassan M, Arpa G, Neri G, Luinetti O, Lenti MV, Ulivi P, Tedaldi G, Furlan D, Quaquarini E, Ardizzone S, Sampietro G, Biancone L, Monteleone G, Solcia E, Sessa F, Paulli M, Adsay NV, Di Sabatino A. Poorly Cohesive Carcinoma of the Nonampullary Small Intestine: A Distinct Histologic Subtype With Prognostic Significance. Am J Surg Pathol 2022; 46:498-508. [PMID: 34628432 DOI: 10.1097/pas.0000000000001821] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Poorly cohesive carcinomas (PCCs) are neoplasms characterized by a dyshesive cell invasion pattern featuring single-cell or cord-like stromal infiltration. Although they have been extensively studied in the stomach and other digestive system organs, limited data regarding nonampullary small bowel poorly cohesive carcinomas (SB-PCCs) are hitherto available. The aims of our study were to analyze the clinicopathologic and immunophenotypical features of SB-PCCs (PCC pattern accounting for >50% of the neoplasm) and to compare them with small bowel adenocarcinomas (SBAs), not otherwise specified (SBAs-NOS) and with cancers with a histologically distinct PCC component accounting for 10% to 50% of the neoplasm (mixed-poorly-cohesive-glandular-SBAs). Fifteen SB-PCCs were identified and compared with 95 SBAs-NOS and 27 mixed-poorly-cohesive-glandular-SBAs. Most SB-PCCs (67%) were composed of <10% of signet-ring cells, and all but 1 SB-PCCs exhibited loss of membranous expression of E-cadherin. Compared with SBAs-NOS, SB-PCCs showed a significantly younger patient age at diagnosis, and a stronger association with Crohn disease, and both SB-PCCs and mixed-poorly-cohesive-glandular-SBAs featured a higher rate of lymphovascular and perineural invasion and a lower percentage of mismatch repair-deficient cases. Importantly, the cancer-specific survival of SB-PCC (hazard ratio: 3.81; 95% confidence interval: 1.90-7.64; P<0.001) and mixed-poorly-cohesive-glandular-SBA (4.12; 2.20-7.71; P<0.001) patients was significantly worse compared with SBAs-NOS cases. This study provides objective evidence to the World Health Organization (WHO) 2019 introduction of SB-PCC as a distinctive subtype of nonampullary SBA, by virtue of its unique clinical and histologic features, and suggests that both SB-PCCs and mixed-poorly-cohesive-glandular-SBAs should be separated from SBAs-NOS.
Collapse
Affiliation(s)
- Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
- Anatomic Pathology, Fondazione IRCCS San Matteo Hospital
| | - Camilla Guerini
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
| | - Federica Grillo
- Pathology Unit, Department of Surgical and Diagnostic Sciences (DISC), University of Genoa and Ospedale Policlinico San Martino IRCCS, Genoa
| | - Catherine Klersy
- Clinical Epidemiology & Biometry Unit, Fondazione IRCCS San Matteo Hospital
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua
- Veneto Institute of Oncology, IOV-IRCCS, Padua
| | - Giovanni Arpa
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
| | - Giuseppe Neri
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
| | | | - Marco V Lenti
- Department of Internal Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola
| | - Gianluca Tedaldi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola
| | - Daniela Furlan
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese
| | | | - Sandro Ardizzone
- Gatroenterology Unit, ASST Fatebenefratelli-Sacco, L. Sacco University Hospital
- Department of Biochemical and Clinical Sciences, University of Milan, Milan
| | | | - Livia Biancone
- Gatroenterology Unit, Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy
| | - Giovanni Monteleone
- Gatroenterology Unit, Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy
| | - Enrico Solcia
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
| | - Fausto Sessa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese
| | - Marco Paulli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia
- Anatomic Pathology, Fondazione IRCCS San Matteo Hospital
| | - Nazmi V Adsay
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Antonio Di Sabatino
- Department of Internal Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital
| |
Collapse
|
46
|
Pourhoseingholi MA. Epidemiology and burden of gluten-related disorders. GLUTEN-RELATED DISORDERS 2022:59-81. [DOI: 10.1016/b978-0-12-821846-4.00011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
|
48
|
Al-Abachi KT. Screening for Celiac Disease in Patients with Irritable Bowel Syndrome Fulfilling Rome III Criteria. JOURNAL OF COLOPROCTOLOGY 2021. [DOI: 10.1055/s-0041-1736645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Abstract
Background Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder. Celiac disease (CD), a treatable autoimmune enteropathy, with varied presentations, may simulate clinically symptoms of IBS. The aim of the present study is to screen for CD in patients with IBS diagnosed based on the Rome III criteria.
Patients and Methods A cross-sectional study was conducted at a secondary care gastrointestinal unit in Al-Salam General Hospital in Mosul city, Iraq, from November 2015 to October 2016. All patients fulfilling the Rome III criteria for IBS were screened for CD using antitissue transglutaminase IgA antibodies (anti-tTG). Patients who tested positive were subjected to endoscopic duodenal biopsy to confirm the diagnosis of CD.
Results A total of 100 patients were included in the present study (58 female and 42 male), the mean age of the participants was 40.8 years old (standard deviation [SD] ± 11.57). Ten patients (10/100, 10%) tested positive for anti-tTG antibodies. Five of the seropositive patients (5/10, 50%) showed positive biopsy results according to the Marsh classification, 3 of whom having diarrhea, and 2 with constipation.
Conclusion Positive serology and biopsy results suggestive of CD are common among patients with IBS. Screening patients with IBS for CD is justified.
Collapse
|
49
|
Lauxmann MA, Vazquez DS, Schilbert HM, Neubauer PR, Lammers KM, Dodero VI. From celiac disease to coccidia infection and vice-versa: The polyQ peptide CXCR3-interaction axis. Bioessays 2021; 43:e2100101. [PMID: 34705290 DOI: 10.1002/bies.202100101] [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: 04/13/2021] [Revised: 09/04/2021] [Accepted: 10/06/2021] [Indexed: 11/11/2022]
Abstract
Zonulin is a physiological modulator of intercellular tight junctions, which upregulation is involved in several diseases like celiac disease (CeD). The polyQ gliadin fragment binds to the CXCR3 chemokine receptor that activates zonulin upregulation, leading to increased intestinal permeability in humans. Here, we report a general hypothesis based on the structural connection between the polyQ sequence of the immunogenic CeD protein, gliadin, and enteric coccidian parasites proteins. Firstly, a novel interaction pathway between the parasites and the host is described based on the structural similarities between polyQ gliadin fragments and the parasite proteins. Secondly, a potential connection between coccidial infections as a novel environmental trigger of CeD is hypothesized. Therefore, this report represents a promising breakthrough for coccidian research and points out the potential role of coccidian parasites as a novel trigger of CeD that might define a preventive strategy for gluten-related disorders in general. Also see the video abstract here: https://youtu.be/oMaQasStcFI.
Collapse
Affiliation(s)
- Martin A Lauxmann
- Institute for Biochemistry, Brandenburg Medical School (MHB) Theodor Fontane, Germany.,Department of Nephrology, Campus Clinic Brandenburg, Brandenburg Medical School (MHB) Theodor Fontane, Germany
| | - Diego S Vazquez
- Grupo de Biología Estructural y Biotecnología (GBEyB-IMBICE), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Hanna M Schilbert
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany.,Genetics and Genomics of Plants, Center for Biotechnology (CeBiTec) & Faculty of Biology, Universitätsstraße 25, Bielefeld, 33615, Germany
| | - Pia R Neubauer
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany
| | | | - Veronica I Dodero
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany
| |
Collapse
|
50
|
Kiliccalan I. Is the Rotavirus Vaccine Really Associated with a Decreased Risk of Developing Celiac and Other Autoimmune Diseases? Rambam Maimonides Med J 2021; 12:RMMJ.10450. [PMID: 34449304 PMCID: PMC8549836 DOI: 10.5041/rmmj.10450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review examines the risk of developing celiac disease (CD) and other autoimmune diseases in individuals receiving the rotavirus (RV) vaccine compared to the normal population. Celiac disease is a malabsorptive, chronic, immune-mediated enteropathy involving the small intestine. The pathogenesis of CD is multifactorial, and mucosal immunity plays an important role in its development. Low mucosal IgA levels significantly increase the risk of developing the disease. Rotavirus is an infectious agent that causes diarrhea, particularly in children aged 0-24 months, and is frequently involved in diarrhea-related deaths in these children. An oral vaccine against RV has been developed. While it is effective on RV infection, it also contributes to increasing mucosal immunity. Studies have indicated that individuals immunized with the RV vaccine are at lower risk of developing CD than unvaccinated individuals. In addition, the mean age for developing CD autoimmunity may be higher in the vaccinated group than in controls receiving placebo. Additional studies that include children immunized with different RV vaccines and unvaccinated children would provide more meaningful results. Although current data suggest a possible association of RV vaccination with a reduced risk of developing CD and other autoimmune diseases, this remains an unanswered question that merits greater international investigation.
Collapse
|