1
|
Ludvigsson JF, Yao J, Lebwohl B, Green PHR, Yuan S, Leffler DA. Coeliac disease: complications and comorbidities. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-024-01032-w. [PMID: 39875649 DOI: 10.1038/s41575-024-01032-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Coeliac disease is an autoimmune disease characterized by small intestinal villus atrophy and inflammation upon exposure to gluten. It has a global prevalence of approximately 1%. Although the gluten-free diet can be an effective treatment, this diet is burdensome with practical difficulties and frequent inadvertent gluten exposure. Moreover, there are a variety of potential complications and comorbidities of coeliac disease that might be related to malabsorption and/or chronic immune activation. Overall, individuals with coeliac disease have increased mortality compared with the general population, underscoring the severity of this common disease. Comorbidities and complications that have been associated with coeliac disease include poor growth, reproductive complications, kidney and liver diseases, respiratory disease (such as pneumonia) and infections (including sepsis). Furthermore, coeliac disease has been linked to other autoimmune disease and psychiatric disease, as well as certain cancers. Data suggest that mucosal healing on a gluten-free diet might protect against some, but not all, of these complications. In this Review, we present absolute and relative risks of coeliac-associated disorders. We discuss underlying mechanisms, the role of the gluten-free diet and mucosal healing, as well as implications for follow-up and non-dietary treatment of coeliac disease.
Collapse
Affiliation(s)
- Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Celiac Disease Center, Department of Medicine, Columbia University Medical Center, New York, NY, USA.
- Department of Paediatrics, Orebro University Hospital, Orebro, Sweden.
| | - Jialu Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Lebwohl
- Celiac Disease Center, Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Peter H R Green
- Celiac Disease Center, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel A Leffler
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Celiac Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Santonicola A, Soldaini C, Ciacci C. New therapies in celiac disease. Curr Opin Gastroenterol 2025:00001574-990000000-00176. [PMID: 39862215 DOI: 10.1097/mog.0000000000001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
PURPOSE OF REVIEW Celiac disease (CeD) is a chronic autoimmune disorder of the small intestine triggered by gluten ingestion in genetically predisposed individuals. The cornerstone of CeD management remains a strict adherence to a lifelong gluten-free diet (GFD), although such a dietary restriction can lead to an altered quality of life and may not be easy to follow for many patients. These challenges highlighted the need for alternative therapies. This review aims to explore the latest advancements in these therapeutic avenues, emphasizing mechanisms of action, clinical efficacy, and safety profiles of drugs currently in advanced stages of clinical testing. RECENT FINDINGS Recent advances in the understanding of CeD pathophysiology have catalyzed the development of new therapeutic approaches, which include strategies to modify gluten processing in the gut, block gluten-triggered immune responses, or restore immune tolerance to gluten. SUMMARY While these therapies are not poised to take the place of GFD, they represent promising treatment alternatives that could enhance the quality of life and minimize long-term consequences in CeD patients. Further research, as well as phase III clinical trials of those already conducted, are needed to establish the feasibility of integrating these novel drugs in the clinical management of CeD.
Collapse
Affiliation(s)
- Antonella Santonicola
- Gastrointestinal Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | | | | |
Collapse
|
3
|
Cheung T, McDonald C, Setty M, Tsai P, Wadhwani SI. Social Adversities Associate with Worse Disease Control in Pediatric Celiac Disease. J Pediatr 2025; 276:114305. [PMID: 39293560 PMCID: PMC11812434 DOI: 10.1016/j.jpeds.2024.114305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/24/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE To characterize how social adversities influence disease control in children with celiac disease (CeD). STUDY DESIGN We conducted a cross-sectional analysis of data from 325 eligible children ≤18 years old with CeD enrolled between 2015 through 2023 into iCureCeliac, a patient-centered US registry for CeD. We evaluated the associations between financial insecurity, social stigmatization, decreased health knowledge, and mental health comorbidity with 2 validated patient-reported outcomes on disease activity and gluten-free diet adherence: celiac symptom index and CeD adherence test, respectively. We used multivariable logistic and linear regression analysis to adjust for race, primary spoken language, and socioeconomic status. RESULTS Among 325 children with available financial insecurity data, the median age was 11 years (IQR 8, 15), 67% were female, and 88% were White. In multivariable logistic regression, the odds of elevated disease activity among children with financial insecurity, social stigmatization, decreased health knowledge, and mental health comorbidity were 2.6 (95% CI 0.9, 8.0; P = .09), 2.8 (95% CI 1.6, 5.1; P < .001), 4.8 (95% CI 2.4, 9.8; P < .001), and 1.9 (95% CI 1.1, 3.3; P = .03), respectively. For insufficient dietary adherence, the respective odds were 1.6 (95% CI 0.5, 4.7; P = .43), 3.3 (95% CI 1.7, 6.5; P < .001), 2.9 (95% CI 1.5, 5.7; P = .002), and 2.3 (95% CI 1.2, 4.2; P = .01). Statistically significant associations in logistic regression aligned with results of linear models. CONCLUSIONS Social stigmatization, decreased health knowledge, and mental health comorbidity were associated with worse disease control in pediatric CeD. Targeted interventions aimed at addressing these social adversities may improve disease activity and dietary adherence.
Collapse
Affiliation(s)
- Telly Cheung
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Christine McDonald
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Mala Setty
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Patrika Tsai
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Sharad I Wadhwani
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA.
| |
Collapse
|
4
|
Wei Y, Wang Y, Yuan Y, Chen J. Celiac Disease, Gluten-Free Diet, and Eating Disorders: From Bench to Bedside. Foods 2024; 14:74. [PMID: 39796364 PMCID: PMC11720414 DOI: 10.3390/foods14010074] [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: 11/08/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Celiac disease (CD) and eating disorders (EDs) are complex chronic conditions in adolescents, sharing symptoms such as weight change, malnutrition, and gastrointestinal symptoms. CD, an autoimmune disorder triggered by gluten ingestion, is managed through a strict gluten-free diet that can unintentionally foster disordered eating behaviors due to dietary restrictions. Conversely, EDs may mask and complicate CD symptoms, leading to diagnostic delays and treatment challenges. Evidence reveals an increased risk of EDs in CD individuals and vice versa, indicating a potential bidirectional relationship. This review explores the mechanisms and clinical implications of this interplay and proposes integrated screening and care strategies to improve the quality of life for individuals with both conditions.
Collapse
Affiliation(s)
- Yaohui Wei
- Department of Clinical Psychology, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Y.W.); (Y.W.)
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Yating Wang
- Department of Clinical Psychology, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Y.W.); (Y.W.)
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Jue Chen
- Department of Clinical Psychology, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Y.W.); (Y.W.)
| |
Collapse
|
5
|
Ciaccio EJ, Lee AR, Lebovits J, Wolf RL, Lewis SK. Physical and psychological symptoms and survey importance in celiac disease. World J Gastrointest Endosc 2024; 16:632-639. [PMID: 39735391 PMCID: PMC11669964 DOI: 10.4253/wjge.v16.i12.632] [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: 05/24/2024] [Revised: 09/20/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Celiac disease is an autoimmune condition that affects approximately 1% of the worldwide community. Originally thought to be confined mostly to the small intestine, resulting in villous atrophy and nutrient malabsorption, it has more recently been implicated in systemic manifestations as well, particularly when undiagnosed or left untreated. Herein, the physical and psychological symptoms of celiac disease are described and explored. An emphasis is placed on efforts to query prospective and confirmed celiac disease patients via the use of surveys. Suggestions are made regarding the development of efficacious surveys for the purpose of screening for celiac disease in undiagnosed persons, and monitoring efficacy of the gluten-free diet in persons diagnosed with celiac disease. There are broad categories of physical and psychological symptoms associated with celiac disease. There is also an essential interaction between such physical and the psychological symptoms. It is important to capture the association between symptoms, via queries directed toward suspected and confirmed persons with celiac disease. The use of anonymous online surveys can be helpful to determine the qualities and characteristics which may be associated with this condition. It is suggested that personal surveys should be given a greater role in screening and to lessen the time for diagnosis. Querying the subject directly via a survey can provide clues as to the types of symptoms being experienced by those with celiac disease currently, as well as to determine the salient aspects of the symptomatology, which will be useful for rapid screening and monitoring in future work.
Collapse
Affiliation(s)
- Edward J Ciaccio
- Celiac Disease Center at Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| | - Anne R Lee
- Celiac Disease Center at Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| | - Jessica Lebovits
- Celiac Disease Center at Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| | - Randi L Wolf
- Department of Health Studies and Applied Educational Psychology, Columbia University, Teachers College, New York, NY 10027, United States
| | - Suzanne K Lewis
- Celiac Disease Center at Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| |
Collapse
|
6
|
Howard K, Adelman D, Ghura S, Acaster S, Clifford S, Kelly CP, Martin SA, Meckley LM, Leffler DA. Development of the celiac disease symptom diary version 2.1 © (CDSD 2.1 ©) patient-reported outcome measure. Qual Life Res 2024; 33:3275-3282. [PMID: 39461929 PMCID: PMC11599373 DOI: 10.1007/s11136-024-03799-6] [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] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
PURPOSE For patients with celiac disease (CeD), the only current management option is adherence to a strict gluten-free diet (GFD); however, many patients on a GFD continue to experience symptoms with a significant impact on quality of life. Potential new treatments for CeD are under development and a validated patient-reported outcome measure is required to evaluate their utility in clinical trials. The purpose of this article is to provide a history of the development of the Celiac Disease Symptom Diary (CDSD) 2.1© for use in clinical trials. METHODS Qualitative and quantitative studies were conducted from 2010 to 2021, including concept elicitation and cognitive debriefing interviews with adult and adolescent participants with CeD (N = 93) diagnosed via biopsy and/or serology and input from eight interviews with CeD clinical experts. During these studies, different iterations of the CDSD were presented to the US Food and Drug Administration and the European Medicines Agency, and modifications were made in line with their feedback. RESULTS These studies ultimately led to the development of CDSD 2.1©, a daily diary which focuses on key symptoms of CeD (abdominal pain, bloating, diarrhea, nausea and tiredness). This patient-reported outcome measure was readily understood by adult and adolescent participants with CeD and content validity was demonstrated in both populations. CONCLUSION CDSD 2.1© is a content-valid patient-reported outcome measure developed in accordance with best practices and regulatory guidance. A thorough exploration of the psychometric properties of CDSD 2.1© for both adult and adolescent participants with CeD is ongoing to support utilization in clinical trials.
Collapse
Affiliation(s)
- Kellee Howard
- ICON plc, San Fransisco, CA, USA
- Patient Centered Solutions, IQVIA, Montreal, QC, Canada
| | | | - Sonal Ghura
- Takeda Development Center Americas Inc, Cambridge, CA, USA
| | - Sarah Acaster
- Oxford Outcomes Ltd. (later acquired by ICON plc), Oxford, UK
- Acaster Lloyd Consulting Ltd., London, UK
| | - Sarah Clifford
- ICON plc, San Fransisco, CA, USA
- Sprout Health Solutions, Los Angeles, CA, USA
| | - Ciaran P Kelly
- Celiac Center, Beth Israel Deaconess Medical Center, Harvard Medical School Celiac Research Program, Boston, MA, USA
| | | | - Lisa M Meckley
- Takeda Development Center Americas Inc., Cambridge, MA, USA
| | - Daniel A Leffler
- Takeda Development Center Americas Inc, Cambridge, CA, USA.
- Celiac Center, Beth Israel Deaconess Medical Center, Harvard Medical School Celiac Research Program, Boston, MA, USA.
| |
Collapse
|
7
|
Roque A, Zanker J, Brígido S, Tomaz MB, Gonçalves A, Barbeiro S, Benítez-Páez A, Pereira SG. Dietary patterns drive loss of fiber-foraging species in the celiac disease patients gut microbiota compared to first-degree relatives. Gut Pathog 2024; 16:58. [PMID: 39375796 PMCID: PMC11459851 DOI: 10.1186/s13099-024-00643-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Celiac disease is an autoimmune disorder triggered by dietary gluten in genetically predisposed individuals that primarily affects the small intestine. Studies have reported differentially abundant bacterial taxa in the gut microbiota of celiac patients compared with non-celiac controls. However, findings across studies have inconsistencies and no microbial signature of celiac disease has been defined so far. RESULTS Here, we showed, by comparing celiac patients with their non-celiac 1st-degree relatives, that bacterial communities of related individuals have similar species occurrence and abundance compared with non-relatives, regardless the disease status. We also found in celiac patients a loss of bacterial species associated with fiber degradation, and host metabolic and immune modulation, as ruminiclostridia, ruminococci, Prevotella, and Akkermansia muciniphila species. We demonstrated that the differential abundance of bacterial species correlates to different dietary patterns observed between the two groups. For instance, Ruminiclostridium siraeum, Ruminococcus bicirculans, and Bacteroides plebeious, recognized as fiber-degraders, appear more abundant in non-celiac 1st-degree relatives, which have a vegetable consumption pattern higher than celiac patients. Pattern of servings per day also suggests a possible link between these species' abundance and daily calorie intake. CONCLUSIONS Overall, we evidenced that a kinship approach could be valuable in unveiling potential celiac disease microbial traits, as well as the significance of dietary factors in shaping microbial profiles and their influence on disease development and progression. Our results pave the way for designing and adopting novel dietary strategies based on gluten-free fiber-enriched ingredients to improve disease management and patients' quality of life.
Collapse
Affiliation(s)
- Ana Roque
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal
| | - Joyce Zanker
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal
| | - Sara Brígido
- Labeto, Centro de Análises Bioquímicas SA, Beatriz Godinho Laboratories, 2410-152, Leiria, Portugal
| | - Maria Beatriz Tomaz
- Labeto, Centro de Análises Bioquímicas SA, Beatriz Godinho Laboratories, 2410-152, Leiria, Portugal
| | - André Gonçalves
- Gastroentherology Division, Leiria Central Hospital, Leiria Region Local Health Unit, 2410-197, Leiria, Portugal
| | - Sandra Barbeiro
- Gastroentherology Division, Leiria Central Hospital, Leiria Region Local Health Unit, 2410-197, Leiria, Portugal
| | - Alfonso Benítez-Páez
- Microbiome, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980, Paterna, Valencia, Spain
| | - Sónia Gonçalves Pereira
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal.
| |
Collapse
|
8
|
Ge HJ, Chen XL. Advances in understanding and managing celiac disease: Pathophysiology and treatment strategies. World J Gastroenterol 2024; 30:3932-3941. [PMID: 39351055 PMCID: PMC11438662 DOI: 10.3748/wjg.v30.i35.3932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
In this editorial, we comment on an article published in the recent issue of the World Journal of Gastroenterology. Celiac disease (CeD) is a disease occurring in genetically susceptible individuals, which is mainly characterized by gluten intolerance in the small intestine and clinical symptoms such as abdominal pain, diarrhea, and malnutrition. Therefore, patients often need a lifelong gluten-free diet, which greatly affects the quality of life and expenses of patients. The gold standard for diagnosis is intestinal mucosal biopsy, combined with serological and genetic tests. At present, the lack of safe, effective, and satisfactory drugs for CeD is mainly due to the complexity of its pathogenesis, and it is difficult to find a perfect target to solve the multi-level needs of patients. In this editorial, we mainly review the pathological mechanism of CeD and describe the current experimental and improved drugs for various pathological aspects.
Collapse
Affiliation(s)
- Hao-Jie Ge
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
9
|
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
|
10
|
Buriánek F, Gege C, Marinković P. New developments in celiac disease treatments. Drug Discov Today 2024; 29:104113. [PMID: 39067614 DOI: 10.1016/j.drudis.2024.104113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Celiac disease (CeD), an autoimmune disorder triggered by gluten, affects around 1% of the global population. Standard treatment is a strict gluten-free diet (GFD), which poses significant challenges due to dietary restrictions, cross-contamination and subsequent persistent intestinal inflammation. This underscores the need for new treatment options addressing the complex pathophysiology of CeD. Recent research focuses on developing drugs that target intestinal barrier regeneration, gluten peptide modification, immune response alteration, and gut microbial ecosystem modulation. These approaches offer potential for more effective management of CeD beyond GFD. Gluten-independent treatments may be particularly relevant under the FDA's draft guidance for CeD, which emphasizes drug development as an adjunct to GFD for patients with ongoing signs and symptoms of CeD despite strict GFD.
Collapse
|
11
|
Angyal MM, Janssen MF, Lakatos PL, Brodszky V, Rencz F. The added value of the cognition, dining, gastrointestinal problems, sleep and tiredness bolt-on dimensions to the EQ-5D-5L in patients with coeliac disease. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2024:10.1007/s10198-024-01719-6. [PMID: 39212881 DOI: 10.1007/s10198-024-01719-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Multiple studies suggest that the EQ-5D may overestimate health-related quality of life (HRQoL) in patients with coeliac disease (CD). We aimed to develop and psychometrically test potentially relevant bolt-on dimensions to improve the measurement performance of the EQ-5D-5L in CD patients. METHODS The development and selection of bolt-ons were informed by a literature review on HRQoL in CD, expert and patient input. A cross-sectional online survey was conducted amongst 312 adult CD patients. Respondents completed the EQ-5D-5L, two condition-specific bolt-ons newly-developed for the present study [dining (DI) and gastrointestinal problems (GI)] and three existing bolt-ons [cognition (CO), sleep (SL) and tiredness (TI)]. The following psychometric properties were tested: ceiling, informativity, convergent and known-group validity, and dimensionality (confirmatory factor analysis). RESULTS Adding the TI, SL, GI, DI and CO individual bolt-ons reduced the ceiling of the EQ-5D-5L (39%) to 17%, 23%, 24%, 26% and 37%, respectively. GI excelled with strong convergent validity with the Gastrointestinal Symptom Rating Scale total score (rs=0.71) and improved the discriminatory power for all known-groups. GI was the only bolt-on loading on a different factor from the five core dimensions, whereas the other four bolt-ons loaded onto the same 'psychosocial health' factor as the EQ-5D-5L anxiety/depression dimension. CONCLUSION The DI, GI, SL and TI bolt-ons, especially the GI, enhance the validity of EQ-5D-5L in patients with CD, suggesting their value in capturing important HRQoL aspects potentially missed by the five core dimensions. These bolt-ons can be used in sensitivity analyses supporting health technology assessments and subsequent resource allocation decisions.
Collapse
Affiliation(s)
- M Mercédesz Angyal
- Károly Rácz Conservative Medicine Division, Semmelweis University Doctoral School, 26 Üllői út, Budapest, H-1085, Hungary
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary
| | - Mathieu F Janssen
- Section Medical Psychology and Psychotherapy, Department of Psychiatry, Erasmus MC, Rotterdam, The Netherlands
| | - Péter L Lakatos
- McGill University Health Centre, Montreal General Hospital, 1650 Ave. Cedar, D16.173.1, Montreal, QC, H3G 1A4, Canada
- Department of Internal Medicine and Oncology, Semmelweis University, 2/a Korányi Sándor utca, Budapest, 1083, Hungary
| | - Valentin Brodszky
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary
| | - Fanni Rencz
- Károly Rácz Conservative Medicine Division, Semmelweis University Doctoral School, 26 Üllői út, Budapest, H-1085, Hungary.
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary.
| |
Collapse
|
12
|
Riznik P, Carnohorski I, Dolinsek J, Dragutinovic N, Gyimesi J, Hauer AC, Klemenak M, Korponay-Szabo IR, Krencnik T, Masic M, Misak Z, Pavkov V, Popp A, Raba T, Szitanyi P, Dolinsek J. Uncovering the gap: Coeliac disease knowledge among healthcare professionals in the Danube region. BMC Gastroenterol 2024; 24:254. [PMID: 39123100 PMCID: PMC11312701 DOI: 10.1186/s12876-024-03349-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Several studies have shown that the knowledge about coeliac disease (CD) is not satisfactory among healthcare professionals (HCP). The aim of our study was to assess the knowledge of HCPs about CD in the Danube region. METHODS HCPs from 8 countries in the Danube region were asked to complete the web-based questionnaire about CD. Scores of HCPs were compared according to their speciality, work experience and country of residence. The results were compared with the results of a similar study conducted in Central Europe within the Focus IN CD project in 2016. RESULTS Questionnaire was completed by 799 HCPs from Austria, Croatia, Czech Republic, Hungary, Moldova, Romania, Serbia, and Slovenia. Mean score achieved by HCPs was 52.2%. Paediatric gastroenterologists scored the highest (75.3%). Comparing the data with the study conducted in Central Europe in 2016, we found a significant rise (p < 0.001) in the knowledge of paediatric gastroenterologists. Also, HCPs who previously took part in the Focus IN CD project, achieved higher score (61.1% vs. 50.8%; p < 0.001). CONCLUSION The knowledge about CD among HCPs in Danube region is not satisfactory. There has been a significant increase in the knowledge of paediatric gastroenterologists, showing the benefit of various awareness raising activities that were carried out recently.
Collapse
Affiliation(s)
- Petra Riznik
- Paediatric Department, Gastroenterology, Hepatology and Nutrition Unit, University Medical Centre Maribor, Maribor, Slovenia.
| | | | - Jasmina Dolinsek
- Municipality of Maribor, Project development office, Maribor, Slovenia
| | - Natasa Dragutinovic
- Department of Gastroenterology, Hepatology and Nutritional Disorders, University Children's Hospital, Belgrade, Serbia
| | - Judit Gyimesi
- Coeliac Disease Centre, Heim Pal National Paediatric Institute, Budapest, Hungary
| | - Almuthe Christine Hauer
- Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Martina Klemenak
- Paediatric Department, Gastroenterology, Hepatology and Nutrition Unit, University Medical Centre Maribor, Maribor, Slovenia
| | - Ilma Rita Korponay-Szabo
- Coeliac Disease Centre, Heim Pal National Paediatric Institute, Budapest, Hungary
- Medical Faculty, Department of Paediatrics, Coeliac Disease Study Group, University of Debrecen, Debrecen, Hungary
| | - Tomaz Krencnik
- Paediatric Department, Gastroenterology, Hepatology and Nutrition Unit, University Medical Centre Maribor, Maribor, Slovenia
| | - Mario Masic
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Zrinjka Misak
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | | | - Alina Popp
- Department of Paediatrics, University of Medicine and Pharmacy and National Institute for Mother and Child Health Alessandrescu-Rusescu, Bucharest, Romania
| | - Tatiana Raba
- Paediatric Department, "Nicolae Testemitanu" State University of Medicine and Pharmacy of the Republic of Moldova, Chisinau, Republic of Moldova
| | - Peter Szitanyi
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, General University Hospital in Prague, Prague, Czech Republic
| | - Jernej Dolinsek
- Paediatric Department, Gastroenterology, Hepatology and Nutrition Unit, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, Paediatric Department, University of Maribor, Maribor, Slovenia
| |
Collapse
|
13
|
Santos AJM, van Unen V, Lin Z, Chirieleison SM, Ha N, Batish A, Chan JE, Cedano J, Zhang ET, Mu Q, Guh-Siesel A, Tomaske M, Colburg D, Varma S, Choi SS, Christophersen A, Baghdasaryan A, Yost KE, Karlsson K, Ha A, Li J, Dai H, Sellers ZM, Chang HY, Dunn JCY, Zhang BM, Mellins ED, Sollid LM, Fernandez-Becker NQ, Davis MM, Kuo CJ. A human autoimmune organoid model reveals IL-7 function in coeliac disease. Nature 2024; 632:401-410. [PMID: 39048815 PMCID: PMC11747932 DOI: 10.1038/s41586-024-07716-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
In vitro models of autoimmunity are constrained by an inability to culture affected epithelium alongside the complex tissue-resident immune microenvironment. Coeliac disease (CeD) is an autoimmune disease in which dietary gluten-derived peptides bind to the major histocompatibility complex (MHC) class II human leukocyte antigen molecules (HLA)-DQ2 or HLA-DQ8 to initiate immune-mediated duodenal mucosal injury1-4. Here, we generated air-liquid interface (ALI) duodenal organoids from intact fragments of endoscopic biopsies that preserve epithelium alongside native mesenchyme and tissue-resident immune cells as a unit without requiring reconstitution. The immune diversity of ALI organoids spanned T cells, B and plasma cells, natural killer (NK) cells and myeloid cells, with extensive T-cell and B-cell receptor repertoires. HLA-DQ2.5-restricted gluten peptides selectively instigated epithelial destruction in HLA-DQ2.5-expressing organoids derived from CeD patients, and this was antagonized by blocking MHC-II or NKG2C/D. Gluten epitopes stimulated a CeD organoid immune network response in lymphoid and myeloid subsets alongside anti-transglutaminase 2 (TG2) autoantibody production. Functional studies in CeD organoids revealed that interleukin-7 (IL-7) is a gluten-inducible pathogenic modulator that regulates CD8+ T-cell NKG2C/D expression and is necessary and sufficient for epithelial destruction. Furthermore, endogenous IL-7 was markedly upregulated in patient biopsies from active CeD compared with remission disease from gluten-free diets, predominantly in lamina propria mesenchyme. By preserving the epithelium alongside diverse immune populations, this human in vitro CeD model recapitulates gluten-dependent pathology, enables mechanistic investigation and establishes a proof of principle for the organoid modelling of autoimmunity.
Collapse
MESH Headings
- Humans
- Autoantibodies/immunology
- Autoimmunity
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biopsy
- Celiac Disease/immunology
- Celiac Disease/pathology
- Celiac Disease/metabolism
- Duodenum/immunology
- Duodenum/pathology
- Duodenum/metabolism
- Epitopes/immunology
- Glutens/immunology
- Glutens/metabolism
- GTP-Binding Proteins/metabolism
- GTP-Binding Proteins/immunology
- HLA-DQ Antigens/immunology
- HLA-DQ Antigens/metabolism
- Interleukin-7/metabolism
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Killer Cells, Natural/immunology
- Models, Biological
- Myeloid Cells/immunology
- Organoids/immunology
- Organoids/metabolism
- Organoids/pathology
- Protein Glutamine gamma Glutamyltransferase 2/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- António J M Santos
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent van Unen
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhongqi Lin
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven M Chirieleison
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nhi Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arpit Batish
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua E Chan
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Cedano
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Elisa T Zhang
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qinghui Mu
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Guh-Siesel
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeline Tomaske
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Deana Colburg
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shannon S Choi
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Asbjørn Christophersen
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ani Baghdasaryan
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasper Karlsson
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjie Dai
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Zachary M Sellers
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Department of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bing M Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ludvig M Sollid
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Nielsen Q Fernandez-Becker
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
14
|
Korn LL, Kutyavin VI, Bachtel ND, Medzhitov R. Adverse Food Reactions: Physiological and Ecological Perspectives. Annu Rev Nutr 2024; 44:155-178. [PMID: 38724028 DOI: 10.1146/annurev-nutr-061021-022909] [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] [Indexed: 05/15/2024]
Abstract
While food is essential for survival, it can also cause a variety of harmful effects, ranging from intolerance to specific nutrients to celiac disease and food allergies. In addition to nutrients, foods contain myriads of substances that can have either beneficial or detrimental effects on the animals consuming them. Consequently, all animals evolved defense mechanisms that protect them from harmful food components. These "antitoxin" defenses have some parallels with antimicrobial defenses and operate at a cost to the animal's fitness. These costs outweigh benefits when defense responses are exaggerated or mistargeted, resulting in adverse reactions to foods. Additionally, pathological effects of foods can stem from insufficient defenses, due to unabated toxicity of harmful food components. We discuss the structure of antitoxin defenses and how their failures can lead to a variety of adverse food reactions.
Collapse
Affiliation(s)
- Lisa L Korn
- Department of Medicine, Section of Rheumatology, Allergy, and Clinical Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Nathaniel D Bachtel
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Ruslan Medzhitov
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
15
|
Tye‐Din JA. Evolution in coeliac disease diagnosis and management. JGH Open 2024; 8:e13107. [PMID: 38957478 PMCID: PMC11217771 DOI: 10.1002/jgh3.13107] [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: 03/18/2024] [Revised: 05/04/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
The traditional gut-centric view of coeliac disease is evolving as immune and genetic insights underscore the central importance of a systemic, T cell immune response to gluten in disease pathogenesis. As the field increasingly recognize the limitations of small intestinal histology as the diagnostic standard, data supporting the accuracy of an immune (serologic) diagnosis of coeliac disease - well demonstrated in children - are growing for adults. Novel biomarkers such as interleukin-2 that identify the gluten-specific T cell demonstrate high sensitivity and specificity for coeliac disease and offer the potential for a diagnostic approach that avoids the need for gluten challenge. Asymptomatic disease and manifestations outside the gut pose considerable challenges for diagnosis using a case-finding strategy and enthusiasm for population screening is growing. The gluten-free diet remains a highly restrictive treatment and there is a paucity of controlled data to inform a safe gluten intake threshold. Ongoing symptoms and enteropathy are common and require systematic evaluation. Slowly-responsive disease is prevalent in the older patient diagnosed with coeliac disease, and super-sensitivity to gluten is an emerging concept that may explain many cases of nonresponsive disease. While there is great interest in developing novel therapies for coeliac disease, no drug has yet been registered. Efficacy studies are generally assessing drugs in patients with treated coeliac disease who undergo gluten challenge or in patients with nonresponsive disease; however, substantial questions remain around specific endpoints relevant for patients, clinicians and regulatory agencies and optimal trial design. Novel immune tools are providing informative readouts for clinical trials and are now shaping their design.
Collapse
Affiliation(s)
- Jason A Tye‐Din
- Immunology DivisionWalter and Eliza Hall InstituteParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of GastroenterologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Centre for Food & Allergy ResearchThe Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| |
Collapse
|
16
|
Hardy MY, Henneken LM, Russell AK, Okura Y, Mizoroki A, Ozono Y, Kobayashi S, Murakami Y, Tye-Din JA. A bispecific antibody targeting HLA-DQ2.5-gluten peptides potently blocks gluten-specific T cells induced by gluten ingestion in patients with celiac disease. Clin Immunol 2024; 264:110259. [PMID: 38768856 DOI: 10.1016/j.clim.2024.110259] [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: 04/11/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
The gluten-free diet for celiac disease (CeD) is restrictive and often fails to induce complete symptom and/or mucosal disease remission. Central to CeD pathogenesis is the gluten-specific CD4+ T cell that is restricted by HLA-DQ2.5 in over 85% of CeD patients, making HLA-DQ2.5 an attractive target for suppressing gluten-dependent immunity. Recently, a novel anti-HLA-DQ2.5 antibody that specifically recognizes the complexes of HLA-DQ2.5 and multiple gluten epitopes was developed (DONQ52). OBJECTIVE To assess the ability of DONQ52 to inhibit CeD patient-derived T-cell responses to the most immunogenic gluten peptides that encompass immunodominant T cell epitopes. METHODS We employed an in vivo gluten challenge model in patients with CeD that affords a quantitative readout of disease-relevant gluten-specific T-cell responses. HLA-DQ2.5+ CeD patients consumed food containing wheat, barley, or rye for 3 days with collection of blood before (D1) and 6 days after (D6) commencing the challenge. Peripheral blood mononuclear cells were isolated and assessed in an interferon (IFN)-γ enzyme-linked immunosorbent spot assay (ELISpot) testing responses to gluten peptides encompassing a series of immunodominant T cell epitopes. The inhibitory effect of DONQ52 (4 or 40 μg/mL) was assessed and compared to pan-HLA-DQ blockade (SPVL3 antibody). RESULTS In HLA-DQ2.5+ CeD patients, DONQ52 reduced T cell responses to all wheat gluten peptides to an equivalent or more effective degree than pan-HLA-DQ antibody blockade. It reduced T cell responses to a cocktail of the most immunodominant wheat epitopes by a median of 87% (IQR 72-92). Notably, DONQ52 also substantially reduced T-cell responses to dominant barley hordein and rye secalin derived peptides. DONQ52 had no effect on T-cell responses to non-gluten antigens. CONCLUSION DONQ52 can significantly block HLA-DQ2.5-restricted T cell responses to the most highly immunogenic gluten peptides in CeD. Our findings support in vitro data that DONQ52 displays selectivity and broad cross-reactivity against multiple gluten peptide:HLA-DQ2.5 complexes. This work provides proof-of-concept multi-specific antibody blockade has the potential to meaningfully inhibit pathogenic gluten-specific T-cell responses in CeD and supports ongoing therapeutic development.
Collapse
Affiliation(s)
- M Y Hardy
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - L M Henneken
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - A K Russell
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Y Okura
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Japan
| | - A Mizoroki
- Chugai Pharmabody Research Pty. Ltd., Singapore
| | - Y Ozono
- Research Division, Chugai Pharmaceutical Co., Ltd., Japan
| | - S Kobayashi
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Japan
| | - Y Murakami
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Japan
| | - J A Tye-Din
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia; Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, Victoria, Australia; The Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Saitta D, Henneken LM, Apputhurai P, Chen Yi Mei SL, Tye-Din JA. Budesonide Induces Favourable Histologic and Symptomatic Recovery in Patients with Non-responsive and Refractory Coeliac Disease When Given in an Open Capsule Format. Dig Dis Sci 2024; 69:2548-2557. [PMID: 38683433 PMCID: PMC11258102 DOI: 10.1007/s10620-024-08436-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/10/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Non-responsive coeliac disease (NRCD), where symptoms and enteropathy persist despite a prolonged gluten-free diet (GFD), is common. Refractory coeliac disease (RCD), characterised by malabsorption and extensive enteropathy, is rare but serious. In both, treatment options are limited. Topical budesonide may help and an open capsule format promoting proximal small intestinal delivery may be advantageous. AIM To describe the effect of budesonide and its presentation on mucosal healing, symptoms, and tolerability in NRCD and RCD. METHODS A retrospective cohort study of NRCD and RCD patients who received budesonide for enteropathy despite a strict GFD for over 12 months. Primary outcome was improvement in histology. Symptoms and adverse treatment effects were recorded. RESULTS 50 patients with NRCD (n = 14; 86% F), RCD type 1 (n = 30; 60% F), and RCD type 2 (n = 6 based on aberrant duodenal T cells; 33% F) were identified. Common RCD symptoms were diarrhoea (68%), fatigue (40%), and weight loss (34%). 16 received closed capsule budesonide (CCB) 9 mg OD and 35 open capsule budesonide (OCB) 3 mg 3 times a day. Complete and partial mucosal healing was significantly higher after OCB compared to CCB (p < 0.001, Mann-Whitney U test). Symptom improvement was also significantly higher after OCB compared to CCB (p = 0.002, Mann-Whitney U test). Side effects were mild and self-limiting and were reported in 25% of both cohorts. CONCLUSION OCB was well tolerated and associated with improvements in enteropathy (83%) and symptoms (90%) in NRCD and RCD. Our findings support OCB as the preferred 1st-line therapy for NRCD and RCD type 1.
Collapse
Affiliation(s)
- Daniel Saitta
- Department of Gastroenterology, Western Hospital, Footscray, VIC, Australia
| | - Lee M Henneken
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Pragalathan Apputhurai
- Department of Health Sciences and Biostatistics, Swinburne University of Technology, Melbourne, Australia
| | - Swee Lin Chen Yi Mei
- Department of Gastroenterology, Box Hill Hospital, Box Hill, Victoria, Australia
| | - Jason A Tye-Din
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC, Australia.
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
18
|
Galipeau HJ, Hinterleitner R, Leonard MM, Caminero A. Non-Host Factors Influencing Onset and Severity of Celiac Disease. Gastroenterology 2024; 167:34-50. [PMID: 38286392 PMCID: PMC11653303 DOI: 10.1053/j.gastro.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts; Center for Celiac Research and Treatment, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
19
|
Kurppa K, Mulder CJ, Stordal K, Kaukinen K. Celiac Disease Affects 1% of Global Population: Who Will Manage All These Patients? Gastroenterology 2024; 167:148-158. [PMID: 38290622 DOI: 10.1053/j.gastro.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024]
Abstract
Celiac disease is a common gastrointestinal condition with an estimated global prevalence of up to 1%. Adequate long-term surveillance of patients is imperative to ensure strict adherence to treatment with a gluten-free diet and the ensuing clinical and histologic recovery. Traditionally, this has been accomplished by means of regular on-site attendance at specialist health care facilities, accompanied for most patients by follow-up endoscopic and laboratory tests. However, the rapidly increasing prevalence of celiac disease and the limited health care resources challenge the current centralized and nonindividualized follow-up strategies. The improved noninvasive surveillance tools and online health care services are further changing the landscape of celiac disease management. There is a clear need for more personalized and on-demand follow-up based on early treatment response and patient-related factors associated with long-term prognosis. Additional scientific evidence on the optimal implementation of follow-up for pediatric and adulthood celiac disease is nevertheless called for.
Collapse
Affiliation(s)
- Kalle Kurppa
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Tampere Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland; University Consortium of Seinäjoki, Seinäjoki, Finland.
| | - Chris J Mulder
- Department of Gastroenterology, Amsterdam University Medical Center, Amsterdam, The Netherlands; Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Ketil Stordal
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Katri Kaukinen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
20
|
Schuppan D, Rostami K. Further Progress to Quantify Histological Damage in Patients With Celiac Disease. Clin Gastroenterol Hepatol 2024; 22:1188-1189. [PMID: 38278199 DOI: 10.1016/j.cgh.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Affiliation(s)
- Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, Center for Celiac Disease and Autoimmunity, Johannes-Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, Massachusetts.
| | - Kamran Rostami
- Crest Hospital Specialist Centre, Palmerston North, New Zealand; Department of Gastroenterology MidCentral District Health Board & Crest Specialist Centre, Palmerston North, New Zealand
| |
Collapse
|
21
|
Abadie V, Han AS, Jabri B, Sollid LM. New Insights on Genes, Gluten, and Immunopathogenesis of Celiac Disease. Gastroenterology 2024; 167:4-22. [PMID: 38670280 PMCID: PMC11283582 DOI: 10.1053/j.gastro.2024.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois.
| | - Arnold S Han
- Columbia Center for Translational Immunology, Columbia University, New York, New York; Department of Microbiology and Immunology, Columbia University, New York, New York; Department of Medicine, Digestive and Liver Diseases, Columbia University, New York, New York
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois; Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
22
|
D'heedene M, Vanuytsel T, Wauters L. Celiac disease: Hope for new treatments beyond a gluten-free diet. Clin Nutr 2024; 43:1240-1249. [PMID: 38648685 DOI: 10.1016/j.clnu.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND & AIMS Celiac disease (CD) is a chronic inflammatory disease of the small intestine induced and maintained by gluten ingestion in susceptible individuals. Current treatment consists of strict adherence to a lifelong gluten-free diet (GFD) which is considered safe and effective in the large majority of patients. However, since adherence to a GFD is difficult and has a negative impact on quality of life, an increasing interest in other treatment options has emerged. Moreover, in some individuals a GFD is not sufficiently effective, necessitating alternative treatments. METHODS By performing a systematic search, we constructed a detailed narrative review. Only treatment options considered relevant and conducted in a phase I, II or III clinical trial were included. RESULTS Based on the pathophysiology of CD, four major therapeutic approaches can be distinguished: firstly, by focusing on intraluminal gluten detoxification before absorption occurs, secondly, by modulating intestinal permeability and preventing paracellular uptake, thirdly, by enhancing immunological tolerance to gluten and finally, by regulating gluten auto-immunity. CONCLUSIONS Despite significant efforts, no treatment has yet completed a phase III clinical trial. Future studies will likely focus on the use of supplemental drugs in conjunction to a GFD, with ALV003 and ZED-1227 currently being the most promising therapeutic options.
Collapse
Affiliation(s)
| | - Tim Vanuytsel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat, 49 3000, Leuven, Belgium
| | - Lucas Wauters
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat, 49 3000, Leuven, Belgium.
| |
Collapse
|
23
|
Wan C, Ji T, Wang L, Wu Q, Chen Q, Wang Y, Li Y, He F, Liu W, Zhong W, Wang B. Exploring the molecular mechanisms and shared gene signatures between celiac disease and ulcerative colitis based on bulk RNA and single-cell sequencing: Experimental verification. Int Immunopharmacol 2024; 133:112059. [PMID: 38615385 DOI: 10.1016/j.intimp.2024.112059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/31/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Many immune-mediated diseases have the common genetic basis, as an autoimmune disorder, celiac disease (CeD) primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. As for ulcerative colitis (UC), which most likely involves a complex interplay between some components of the commensal microbiota and other environmental factors in its origin. These two autoimmune diseases share a specific target organ, the bowel. The etiology and immunopathogenesis of both conditions characterized by chronic intestinal inflammation, ulcerative colitis and celiac disease, are not completely understood. Both are complex diseases with genetics and the environmental factors contributing to dysregulation of innate and adaptive immune responses, leading to chronic inflammation and disease. This study is designed to further clarify the relationship between UC and CeD. The GEO database was used to download gene expression profiles for CeD (GSE112102) and UC (GSE75214). The GSEA KEGG pathway analysis revealed that immune-related pathways were significantly associated with both diseases. Further, we screened 187 shared differentially expressed genes (DEGs) of the two diseases. Gene Ontology (GO) and WikiPathways were carried out to perform the biological process and pathway enrichment analysis. Subsequently, based on the DEGs, the least absolute shrinkage and selection operator (LASSO) analysis was performed to screen for the diagnostic biomarkers of the diseases. Moreover, single-cell RNA-sequencing (RNA-seq) data from five colonic propria with UC showed that REG4 expression was present in Goblet cell, Enteroendocrine cell, and Epithelial. Finally, our work identified REG4 is the shared gene of UC and CeD via external data validation, cellular experiments, and immunohistochemistry. In conclusion, our study elucidated that abnormal immune response could be the common pathogenesis of UC and CeD, and REG4 might be a key potential biomarker and therapeutic target for the comorbidity of these two diseases.
Collapse
Affiliation(s)
- Changshan Wan
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Tao Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China; Department of Gastroenterology, Linyi People's Hospital, Shandong 276000, China
| | - Liwei Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Qiuyan Wu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Qiuyu Chen
- Department of Gastroenterology, Tianjin First Central Hospital of Tianjin Medical University, Tianjin 300192, China
| | - Yali Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Yaqian Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Fengming He
- Department of Clinical Laboratory Medicine, Shanxi Medical University, Taiyuan 030600, Shanxi, China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| |
Collapse
|
24
|
Knowles SR, Apputhurai P, Tye-Din JA. Development and validation of a brain fog scale for coeliac disease. Aliment Pharmacol Ther 2024; 59:1260-1270. [PMID: 38445780 DOI: 10.1111/apt.17942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Brain fog is a subjective cognitive impairment commonly reported in coeliac disease. A standardised tool to define and assess it is an important unmet need. AIMS To develop a patient-informed tool to assess brain fog in coeliac disease to support clinical care, research and drug development. METHODS A pilot online study defined patient descriptors of brain fog. A second study evaluated the factor structure and performance of the scale across two-time points ('Now' and in the 'Past week'). One month later, participants were invited to repeat the study with two online cognitive processing tests, the Stroop task and the trail making test. RESULTS Among adults with treated coeliac disease, 37 (91.9% F) participated in the pilot study and 510 (88.8% F) in the second study of whom 99 repeated the study 1 month later with 51 completing cognitive testing. The most common brain fog descriptors were 'difficulty focusing', 'difficulty thinking' and 'difficulty finding the right words and communicating'. The 12-item scale reflects 'cognitive impairment' and 'somatic and affective experience' and demonstrates strong psychometric properties. It tracked with patients report of brain fog being present or absent across two-time points. It did not significantly correlate with the cognitive tests. CONCLUSION The brain fog assessment and severity scale is the first patient-informed clinical outcomes assessment tool measuring brain fog in coeliac disease. It is brief and validated for two time-based formats. Further research coupling it with biomarker discovery is needed to confirm its validity as a predictor of cognitive performance.
Collapse
Affiliation(s)
- Simon R Knowles
- Department of Psychological Sciences, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Australia
| | - Pragalathan Apputhurai
- Department of Health Sciences and Biostatistics, Swinburne University of Technology, Melbourne, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute, Melbourne, Australia
- Department of Gastroenterology, the Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
25
|
Sollid LM. Tolerance-inducing therapies in coeliac disease - mechanisms, progress and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:335-347. [PMID: 38336920 DOI: 10.1038/s41575-024-00895-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Coeliac disease is an autoinflammatory condition caused by immune reactions to cereal gluten proteins. Currently, the only available treatment for the condition is a lifelong avoidance of gluten proteins in the diet. There is an unmet need for alternative therapies. Coeliac disease has a strong association with certain HLA-DQ allotypes (DQ2.5, DQ2.2 and DQ8), and these disease-associated HLA-DQ molecules present deamidated gluten peptides to gluten-specific CD4+ T cells. The gluten-specific CD4+ T cells are the drivers of the immune reactions leading to coeliac disease. Once established, the clonotypes of gluten-specific CD4+ T cells persist for decades, explaining why patients must adhere to a gluten-free diet for life. Given the key pathogenic role of gluten-specific CD4+ T cells, tolerance-inducing therapies that target these T cells are attractive for treatment of the disorder. Lessons learned from coeliac disease might provide clues for treatment of other HLA-associated diseases for which the disease-driving antigens are unknown. Thus, intensive efforts have been and are currently implemented to bring an effective tolerance-inducing therapy for coeliac disease. This Review discusses mechanisms of the various approaches taken, summarizing the progress made, and highlights future directions in this field.
Collapse
Affiliation(s)
- Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
26
|
Jansson-Knodell CL, Rubio-Tapia A. Gluten-related Disorders From Bench to Bedside. Clin Gastroenterol Hepatol 2024; 22:693-704.e1. [PMID: 37879521 DOI: 10.1016/j.cgh.2023.09.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023]
Abstract
Celiac disease, non-celiac gluten sensitivity, and wheat allergy comprise 3 of the main conditions with wheat- and gluten-containing foods as the symptom trigger. Distinguishing between these entities can be daunting. In this review, we compare and contrast celiac disease, non-celiac gluten sensitivity, and wheat allergy to allow clinicians to determine which diagnosis fits their patient to facilitate high-quality management and longitudinal care.
Collapse
Affiliation(s)
- Claire L Jansson-Knodell
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Alberto Rubio-Tapia
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
27
|
Stefanolo JP, Segura V, Grizzuti M, Heredia A, Comino I, Costa AF, Puebla R, Temprano MP, Niveloni SI, de Diego G, Oregui ME, Smecuol EG, de Marzi MC, Verdú EF, Sousa C, Bai JC. Effect of Aspergillus niger prolyl endopeptidase in patients with celiac disease on a long-term gluten-free diet. World J Gastroenterol 2024; 30:1545-1555. [PMID: 38617446 PMCID: PMC11008412 DOI: 10.3748/wjg.v30.i11.1545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/31/2023] [Accepted: 02/18/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND The gluten-free diet (GFD) has limitations, and there is intense research in the development of adjuvant therapies. AIM To examine the effects of orally administered Aspergillus niger prolyl endopeptidase protease (AN-PEP) on inadvertent gluten exposure and symptom prevention in adult celiac disease (CeD) patients following their usual GFD. METHODS This was an exploratory, double-blind, randomized, placebo-controlled trial that enrolled CeD patients on a long-term GFD. After a 4-wk run-in period, patients were randomized to 4 wk of two AN-PEP capsules (GliadinX; AVI Research, LLC, United States) at each of three meals per day or placebo. Outcome endpoints were: (1) Average weekly stool gluten immunogenic peptides (GIP) between the run-in and end of treatments and between AN-PEP and placebo; (2) celiac symptom index (CSI); (3) CeD-specific serology; and (4) quality of life. Stool samples were collected for GIP testing by ELISA every Tuesday and Friday during run-ins and treatments. RESULTS Forty patients were randomized for the intention-to-treat analysis, and three were excluded from the per-protocol assessment. Overall, 628/640 (98.1%) stool samples were collected. GIP was undetectable (< 0.08 μg/g) in 65.6% of samples, and no differences between treatment arms were detected. Only 0.5% of samples had GIP concentrations sufficiently high (> 0.32 μg/g) to potentially cause mucosal damage. Median GIP concentration in the AN-PEP arm was 44.7% lower than in the run-in period. One-third of patients exhibiting GIP > 0.08 μg/g during run-in had lower or undetectable GIP after AN-PEP treatment. Compared with the run- in period, the proportion of symptomatic patients (CSI > 38) in the AN-PEP arm was significantly lower (P < 0.03). AN-PEP did not result in changes in specific serologies. CONCLUSION This exploratory study conducted in a real-life setting revealed high adherence to the GFD. The AN-PEP treatment did not significantly reduce the overall GIP stool concentration. However, given the observation of a significantly lower prevalence of patients with severe symptoms in the AN-PEP arm, further clinical research is warranted.
Collapse
Affiliation(s)
- Juan Pablo Stefanolo
- Small Bowel Section, Department of Medicine, Gastroenterology Hospital of Buenos Aires “Dr. C. Bonorino Udaondo”, Buenos Aires 1264, Argentina
| | - Verónica Segura
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville 41080, Spain
| | - Martina Grizzuti
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires 1264, Argentina
| | - Abel Heredia
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville 41080, Spain
| | - Isabel Comino
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville 41080, Spain
| | - Ana Florencia Costa
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires 1264, Argentina
| | - Roberto Puebla
- Small Bowel Section, Department of Medicine, Gastroenterology Hospital of Buenos Aires “Dr. C. Bonorino Udaondo”, Buenos Aires 1264, Argentina
| | - María Paz Temprano
- Small Bowel Section, Department of Medicine, Gastroenterology Hospital of Buenos Aires “Dr. C. Bonorino Udaondo”, Buenos Aires 1264, Argentina
| | - Sonia Isabel Niveloni
- Small Bowel Section, Department of Medicine, Gastroenterology Hospital of Buenos Aires “Dr. C. Bonorino Udaondo”, Buenos Aires 1264, Argentina
| | - Gabriel de Diego
- Basic and Applied Research Group in Immunology and Bioactives (GIBAIB), Institute of Ecology and Sustainable Development (INEDES), National University of Lujan, Luján 6700, Buenos Aires, Argentina
| | - María E Oregui
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires 1264, Argentina
| | - Edgardo Gustavo Smecuol
- Small Bowel Section, Department of Medicine, Gastroenterology Hospital of Buenos Aires “Dr. C. Bonorino Udaondo”, Buenos Aires 1264, Argentina
| | - Mauricio C de Marzi
- Basic and Applied Research Group in Immunology and Bioactives (GIBAIB), Institute of Ecology and Sustainable Development (INEDES), National University of Lujan, Luján 6700, Buenos Aires, Argentina
| | - Elena F Verdú
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton L8S 4L8, Ontario, Canada
| | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville 41080, Spain
| | - Julio César Bai
- Department of Medicine, Dr. C. Bonorino Udaondo Gastroenterology Hospital, Buenos Aires 1264, Argentina
- Facultad de Medicina, Universidad del Salvador, Buenos Aires C1051ABB, Argentina
| |
Collapse
|
28
|
Roque A, Pereira SG. Bacteria: Potential Make-or-Break Determinants of Celiac Disease. Int J Mol Sci 2024; 25:2090. [PMID: 38396767 PMCID: PMC10889687 DOI: 10.3390/ijms25042090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 02/25/2024] Open
Abstract
Celiac disease is an autoimmune disease triggered by dietary gluten in genetically susceptible individuals that primarily affects the small intestinal mucosa. The sole treatment is a gluten-free diet that places a social and economic burden on patients and fails, in some, to lead to symptomatic or mucosal healing. Thus, an alternative treatment has long been sought after. Clinical studies on celiac disease have shown an association between the presence of certain microbes and disease outcomes. However, the mechanisms that underlie the effects of microbes in celiac disease remain unclear. Recent studies have employed disease models that have provided insights into disease mechanisms possibly mediated by bacteria in celiac disease. Here, we have reviewed the bacteria and related mechanisms identified so far that might protect from or incite the development of celiac disease. Evidence indicates bacteria play a role in celiac disease and it is worth continuing to explore this, particularly since few studies, to the best of our knowledge, have focused on establishing a mechanistic link between bacteria and celiac disease. Uncovering host-microbe interactions and their influence on host responses to gluten may enable the discovery of pathogenic targets and development of new therapeutic or preventive approaches.
Collapse
Affiliation(s)
| | - Sónia Gonçalves Pereira
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541 Leiria, Portugal;
| |
Collapse
|
29
|
Lebwohl B, Ma C, Lagana SM, Pai RK, Baker KA, Zayadi A, Hogan M, Bouma G, Cellier C, Goldsmith JD, Lundin KEA, Pinto-Sanchez MI, Robert ME, Rubio-Tapia A, Sanders DS, Schaeffer DF, Semrad CE, Silvester JA, Verdú EF, Verma R, Wu TT, Feagan BG, Crowley E, Jairath V, Murray JA. Standardizing Randomized Controlled Trials in Celiac Disease: An International Multidisciplinary Appropriateness Study. Gastroenterology 2024; 166:88-102. [PMID: 37704112 DOI: 10.1053/j.gastro.2023.08.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND & AIMS There is a need to develop safe and effective pharmacologic options for the treatment of celiac disease (CeD); however, consensus on the appropriate design and configuration of randomized controlled trials (RCTs) in this population is lacking. METHODS A 2-round modified Research and Development/University of California Los Angeles Appropriateness Method study was conducted. Eighteen gastroenterologists (adult and pediatric) and gastrointestinal pathologists voted on statements pertaining to the configuration of CeD RCTs, inclusion and exclusion criteria, gluten challenge, and trial outcomes. Two RCT designs were considered, representing the following distinct clinical scenarios for which pharmacotherapy may be used: trials incorporating a gluten challenge to simulate exposure; and trials evaluating reversal of histologic changes, despite attempted adherence to a gluten-free diet. Each statement was rated as appropriate, uncertain, or inappropriate, using a 9-point Likert scale. RESULTS For trials evaluating prevention of relapse after gluten challenge, participants adherent to a gluten-free diet for 12 months or more with normal or near-normal-sized villi should be enrolled. Gluten challenge should be FODMAPS (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) free, and efficacy evaluated using histology with a secondary patient-reported outcome measure. For trials evaluating reversal of villus atrophy, the panel voted it appropriate to enroll participants with a baseline villus height to crypt depth ratio ≤2 and measure efficacy using a primary histologic end point. Guidance for measuring histologic, endoscopic, and patient-reported outcomes in adult and pediatric patients with CeD are provided, along with recommendations regarding the merits and limitations of different end points. CONCLUSIONS We developed standardized recommendations for clinical trial design, eligibility criteria, outcome measures, gluten challenge, and disease evaluations for RCTs in patients with CeD.
Collapse
Affiliation(s)
- Benjamin Lebwohl
- Celiac Disease Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York.
| | - Christopher Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Alimentiv Inc, London, Ontario, Canada.
| | - Stephen M Lagana
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Rish K Pai
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, Arizona
| | | | | | | | - Gerd Bouma
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, The Netherlands
| | - Christophe Cellier
- Department of Gastroenterology, University of Paris-Cité, Georges-Pompidou European Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France
| | | | - Knut E A Lundin
- Norwegian Coeliac Disease Research Centre, University of Oslo Faculty of Medicine, Oslo, Norway; Department of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Maria I Pinto-Sanchez
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marie E Robert
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Alberto Rubio-Tapia
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - David S Sanders
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - David F Schaeffer
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Carol E Semrad
- Department of Gastroenterology, University of Chicago, Chicago, Illinois
| | - Jocelyn A Silvester
- Harvard Celiac Research Program, Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Celiac Disease Center, Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Elena F Verdú
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ritu Verma
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, The University of Chicago, Chicago, Illinois
| | - Tsung-Teh Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Brian G Feagan
- Alimentiv Inc, London, Ontario, Canada; Division of Gastroenterology, Department of Medicine, Western University, London, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Eileen Crowley
- Alimentiv Inc, London, Ontario, Canada; Division of Pediatric Gastroenterology, Department of Pediatrics, Children's Hospital Western Ontario, London Health Sciences Centre, Western University, London, Ontario, Canada
| | - Vipul Jairath
- Alimentiv Inc, London, Ontario, Canada; Division of Gastroenterology, Department of Medicine, Western University, London, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
30
|
Chen Y, You Y, Li J, Yang A, Zhou W, Li X. Endoscopic and histopathological hints on infections in patients of common variable immunodeficiency disorder with gastrointestinal symptoms. BMC Gastroenterol 2023; 23:413. [PMID: 38017379 PMCID: PMC10683160 DOI: 10.1186/s12876-023-03052-3] [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: 03/24/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND AND AIMS Common variable immunodeficiency disorder (CVID) patients may have gastrointestinal (GI) involvement and suffer from infections, which are poorly understood. This study aimed to evaluate the clinical, endoscopic, and histopathological features of CVID patients with GI symptoms and determine their correlation with infections. METHODS We performed a retrospective study on 21 CVID patients with GI symptoms who underwent endoscopic examination in Peking Union Medical College Hospital from 2000 to 2020. The clinical, infectious, endoscopic, and histopathological features were reassessed. RESULTS Chronic diarrhea was the most prevalent GI symptom, observed in 95.2% of our CVID cohort. Over 85% of patients had low body weight and malabsorption. Small bowel villous atrophy was found in 90.5% of patients under endoscopy and mostly confirmed by histopathology. GI infections were identified in 9 (42.9%) patients. Of these, 7 patients with diffuse and obvious nodular lymphoid hyperplasia (NLH) of small bowel under endoscopy had significantly higher infection rate (85.7% vs 21.4%, p < 0.05), predominantly with Giardia and bacteria. Small bowel biopsies showed 95% of patients lacked plasma cells and 60% had increased intraepithelial lymphocytes (IELs), but not significantly different between GI infection and non-infection group. Most patients improved after intravenous immunoglobulin and anti-infection therapy. CONCLUSIONS CVID could involve GI tract, particularly small bowel. Obvious NLH under endoscopy could be a hint for GI infection in CVID patients. Comprehensive endoscopic and histopathological evaluation may be helpful in CVID diagnosis and identification of potential co-infection, leading to proper treatment.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yan You
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ji Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Xiaoqing Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
31
|
Lens C, Berne JV, Politis C. The impact of gastrointestinal diseases on oral and maxillofacial surgery outcomes. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 136:577-583. [PMID: 37658034 DOI: 10.1016/j.oooo.2023.05.004] [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: 02/15/2023] [Revised: 03/19/2023] [Accepted: 05/10/2023] [Indexed: 09/03/2023]
Abstract
OBJECTIVE We analyzed the complication rates and outcomes in patients with gastrointestinal disorders who had undergone maxillofacial surgery. STUDY DESIGN We retrospectively analyzed 289 patients with inflammatory bowel disease (IBD), 142 with gastroesophageal reflux disease (GERD), and 42 with celiac disease (452 total) who had undergone oral and/or maxillofacial surgery. We examined their characteristics, medications they had taken, surgeries they had undergone, and complications they had experienced. Based on the results of univariate analysis, we selected variables for multivariable logistic regression to identify independent predictors of postoperative complications. RESULTS We found a complication rate of 10% in patients with IBD, 6.9% in patients with GERD, and 9.5% in patients with celiac disease. Dentoalveolar treatment was significantly associated with an increased risk of postoperative complications in patients with IBD and GERD. CONCLUSIONS Oral and/or maxillofacial surgery is safe in most patients with IBD, GERD, and celiac disease. Preventive measures for postoperative complications should be considered in patients with IBD and GERD undergoing dentoalveolar treatment.
Collapse
Affiliation(s)
- Charlotte Lens
- Oral and Maxillo-facial Surgery-Imaging and Pathology Research Group, Department of Imaging and Pathology, Group Biomedical Sciences, Catholic University Leuven, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.
| | - Jonas Ver Berne
- Oral and Maxillo-facial Surgery-Imaging and Pathology Research Group, Department of Imaging and Pathology, Group Biomedical Sciences, Catholic University Leuven, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Constantinus Politis
- Oral and Maxillo-facial Surgery-Imaging and Pathology Research Group, Department of Imaging and Pathology, Group Biomedical Sciences, Catholic University Leuven, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Xiao B, Zhang C, Zhou J, Wang S, Meng H, Wu M, Zheng Y, Yu R. Design of SC PEP with enhanced stability against pepsin digestion and increased activity by machine learning and structural parameters modeling. Int J Biol Macromol 2023; 250:125933. [PMID: 37482154 DOI: 10.1016/j.ijbiomac.2023.125933] [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: 01/09/2023] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Prolyl endopeptidases from Sphingomonas capsulata (SC PEP) has attracted much attention as promising oral therapy candidate for celiac sprue, however, its low stability in the gastric environment leads to unsatisfactory clinical results. Therefore, improving its stability against pepsin digestion at low pH is crucial for clinical applications, but challenging. In this study, machine learning and physical parameter model were combined to design SC PEP mutants. After iterations, 20 mutants had higher hydrolysis activity in stomach environment, which was up to 14.1-fold compared with wild-type SC PEP. Mutant M24 involving stable and active mutations and pegylated M24 (M24-PEG) had higher activity of hydrolyzing immunogen in bread than wild-type SC PEP in vitro and in vivo, and residual immunogens in simulated gastric environment were only 1/8 and 1/10 of that in the wild-type SC PEP group. The total residual immunogens in the gastrointestinal tract of mice in the M24 and M24-PEG groups were <20 ppm, reaching the standard of non-toxic food. Our results indicate that the combination of M24 (or M24-PEG) with EP-B2 may be a promising candidate for celiac disease, and the strategies developed in this study provide a paradigm for the design of SC PEP stability mutants.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Chun Zhang
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Junxiu Zhou
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Sa Wang
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Huan Meng
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Miao Wu
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Yongxiang Zheng
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China.
| | - Rong Yu
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
33
|
Vauquelin B, Rivière P. [Celiac disease]. Rev Med Interne 2023; 44:539-545. [PMID: 37558601 DOI: 10.1016/j.revmed.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023]
Abstract
Celiac disease is a frequent auto-immune disease characterized by villous atrophy related to gluten intake in patients with genetic susceptiblity. Patients do not present symptoms in the majority of cases. Presence of the disease must be investigated in case of digestive symptoms or presence of auto-immune disease. Diagnosis is based on anti-transglutaminase antibody and dudodenal biospies. The only available treatment is gluten-free diet. Associated auto-immune diseases must be investigated, especially thyroiditis. Complications related to nutritional deficiency must be accounted for also.
Collapse
Affiliation(s)
- B Vauquelin
- Service d'hépato-gastroentérologie et oncologie digestive, centre médico-chirurgical Magellan, CHU de Bordeaux, Bordeaux, France
| | - P Rivière
- Service d'hépato-gastroentérologie et oncologie digestive, centre médico-chirurgical Magellan, CHU de Bordeaux, Bordeaux, France.
| |
Collapse
|
34
|
Castelijn DAR, Mulder AHL, van der Pol P, Hollander JC, Kuiper T, Bijnens C, Damoiseaux J, Bontkes HJ. Multicenter study to compare the diagnostic performance of CLIA vs. FEIA transglutaminase IgA assays for the diagnosis of celiac disease. Clin Chem Lab Med 2023; 61:1446-1454. [PMID: 36857146 DOI: 10.1515/cclm-2022-1045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVES Celiac disease (CD) is an immune-mediated enteropathy driven by gluten intake. Presence of tTG-IgA antibodies is important for the diagnosis. However, different tTG-IgA assays are used and test performance may vary. Therefore, a retrospective multicenter study was performed to compare the diagnostic performance of three assays. METHODS The fluorescence enzyme-linked immunoassay (FEIA) EliA Celikey IgA (Phadia), the chemiluminescence immunoassays (CLIA) h-tTG IgA QUANTA Flash® (Inova Diagnostics) and the anti-tTG ChLIA IgA (Euroimmun) were compared. Diagnostic samples from CD cases (95 adults; 65 children) and controls (479 adults; 253 children) were included. Samples were blinded and reanalyzed on all platforms. RESULTS A high quantitative correlation between platforms was found (p<0.0001). Both CLIA were more sensitive (adults 100%; children 100%) compared to the FEIA (adults 88.4%; children 96.6%). Specificity of all assays was high (≥97.6%) with the FEIA having the highest specificity. A cut-off based on receiver operator characteristic analysis (6.5 U/mL) improved the sensitivity of the FEIA (adults 95.8%; children 100%) without affecting specificity. Cut-off values for the CLIA assays did not need further optimization. With the FEIA, 71% of pediatric cases had a tTG-IgA level ≥10× upper limit of normal compared to 91 and 92% with QUANTA Flash and ChLIA, respectively. CONCLUSIONS All platforms have high diagnostic accuracy. The CLIA assays are more sensitive compared to the FEIA assay. A lower cut-off for the FEIA improves diagnostic performance, particularly in adult cases that, as demonstrated in this study, present with lower tTG-IgA levels compared to pediatric cases.
Collapse
Affiliation(s)
- Daan A R Castelijn
- Laboratory Medical Immunology , Department of Clinical Chemistry , Amsterdam UMC, Location VUMC, The Netherlands; and Amsterdam Institute for Infection and Immunity, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - A H Leontine Mulder
- Clinical Laboratory, Medlon B.V., Enschede, The Netherlands
- Department of Clinical Chemistry, Ziekenhuis Groep Twente, Almelo, The Netherlands
| | - Pieter van der Pol
- Medical Laboratories , Department of Immunology, Reinier Haga MDC, Delft, The Netherlands
| | - Jolien C Hollander
- Laboratory Medical Immunology , Department of Clinical Chemistry , Amsterdam UMC, Location VUMC, The Netherlands; and Amsterdam Institute for Infection and Immunity, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Tietie Kuiper
- Clinical Laboratory, Medlon B.V., Enschede, The Netherlands
| | - Caroline Bijnens
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hetty J Bontkes
- Laboratory Medical Immunology , Department of Clinical Chemistry , Amsterdam UMC, Location VUMC, The Netherlands; and Amsterdam Institute for Infection and Immunity, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Popp A, Laurikka P, Czika D, Kurppa K. The role of gluten challenge in the diagnosis of celiac disease: a review. Expert Rev Gastroenterol Hepatol 2023; 17:691-700. [PMID: 37243608 DOI: 10.1080/17474124.2023.2219893] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/12/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Duodenal biopsy is the gold standard in the diagnosis of celiac disease, with increasing utilization of serology. A gluten challenge may be required, for example, when dietary gluten reduction precedes appropriate diagnostic evaluations. Evidence on the best challenge protocol is currently sparse. Pharmaceutical trials in recent years may have provided new insights into the challenge and advanced the development of novel sensitive histological and immunological methods. AREAS COVERED This review outlines the current perspectives on the use of gluten challenge in the diagnosis of celiac disease and explores future directions in this area. EXPERT OPINION Comprehensive elimination of celiac disease before dietary gluten restriction is essential to avoid diagnostic uncertainties. Gluten challenge continues to have an important role in certain clinical scenarios, although it is important to understand its limitations in the diagnostic evaluation. The evidence so far permits no unequivocal recommendation considering the timing, duration, and amount of gluten used in the challenge. Thus, these decisions should be made on a case-by-case basis. Further studies with more standardized protocols and outcome measures are called for. In the future novel immunological methods may help to shorten or even avoid gluten challenge.
Collapse
Affiliation(s)
- Alina Popp
- Department of Pediatrics, University of Medicine and Pharmacy Carol Davila and National Institute for Mother and Child Health, Bucharest, Romania
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pilvi Laurikka
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Diana Czika
- Department of Pediatrics, University of Medicine and Pharmacy Carol Davila and National Institute for Mother and Child Health, Bucharest, Romania
| | - Kalle Kurppa
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
- The University Consortium of Seinäjoki, Seinäjoki, Finland
| |
Collapse
|
36
|
Isola J, Mäki M, Hils M, Pasternack R, Viiri K, Dotsenko V, Montonen T, Zimmermann T, Mohrbacher R, Greinwald R, Schuppan D. The Oral Transglutaminase 2 Inhibitor ZED1227 Accumulates in the Villous Enterocytes in Celiac Disease Patients during Gluten Challenge and Drug Treatment. Int J Mol Sci 2023; 24:10815. [PMID: 37445994 DOI: 10.3390/ijms241310815] [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: 05/16/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The enzyme transglutaminase 2 (TG2) plays a key role in celiac disease (CeD) pathogenesis. Active TG2 is located mainly extracellularly in the lamina propria but also in the villous enterocytes of the duodenum. The TG2 inhibitor ZED1227 is a promising drug candidate for treating CeD and is designed to block the TG2-catalyzed deamidation and crosslinking of gliadin peptides. Our aim was to study the accumulation of ZED1227 after oral administration of the drug. We studied duodenal biopsies derived from a phase 2a clinical drug trial using an antibody that detects ZED1227 when bound to the catalytic center of TG2. Human epithelial organoids were studied in vitro for the effect of ZED1227 on the activity of TG2 using the 5-biotin-pentylamine assay. The ZED1227-TG2 complex was found mainly in the villous enterocytes in post-treatment biopsies. The signal of ZED1227-TG2 was strongest in the luminal epithelial brush border, while the intensity of the signal in the lamina propria was only ~20% of that in the villous enterocytes. No signal specific to ZED1227 could be detected in pretreatment biopsies or in biopsies from patients randomized to the placebo treatment arm. ZED1227-TG2 staining co-localized with total TG2 and native and deamidated gliadin peptides on the enterocyte luminal surface. Inhibition of TG2 activity by ZED1227 was demonstrated in epithelial organoids. Our findings suggest that active TG2 is present at the luminal side of the villous epithelium and that inhibition of TG2 activity by ZED1227 occurs already there before gliadin peptides enter the lamina propria.
Collapse
Affiliation(s)
- Jorma Isola
- Jilab Inc., 33520 Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Markku Mäki
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Martin Hils
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany
| | | | - Keijo Viiri
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Valeriia Dotsenko
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Toni Montonen
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | | | | | | | - Detlef Schuppan
- Institute of Translational Immunology and Celiac Center, Medical Center, Johannes-Gutenberg University, 55099 Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Angyal MM, Lakatos PL, Jenei B, Brodszky V, Rencz F. Health utilities and willingness to pay in adult patients with coeliac disease in Hungary. Qual Life Res 2023:10.1007/s11136-023-03418-w. [PMID: 37067657 PMCID: PMC10393904 DOI: 10.1007/s11136-023-03418-w] [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] [Accepted: 04/05/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Coeliac disease (CD) is a life-long food-related disorder with a global prevalence of approximately 1%. Patients with CD often experience reduced health-related quality of life that could improve with a strict adherence to a gluten-free diet (GFD). OBJECTIVES To obtain visual analogue scale (VAS), time trade-off (TTO) and willingness-to-pay (WTP) values amongst patients with CD. METHODS In 2020-2021, a cross-sectional online survey was conducted amongst 312 adult CD patients in Hungary. Patients completed the Gastrointestinal Symptom Rating Scale (GSRS) and evaluated their current health and three hypothetical health state vignettes defined based on dietary adherence using VAS, conventional 10-year TTO and WTP. Multivariate regressions were used to explore the effect of patients' demographic and clinical characteristics on utility and WTP values. RESULTS Mean VAS values for current health and 'CD with strict adherence to GFD', 'CD with loose adherence to GFD' and 'CD without GFD' hypothetical health states were 79.69 ± 18.52, 85.36 ± 16.18, 62.44 ± 19.91 and 36.69 ± 25.83, respectively. Corresponding mean TTO utilities were: 0.90 ± 0.19, 0.91 ± 0.20, 0.87 ± 0.23 and 0.76 ± 0.29. Mean annual WTP values for returning to full health were: €845 ± 1077, €648 ± 1002, €862 ± 1135 and €1251 ± 1496. Older age at diagnosis, male sex, more severe gastrointestinal symptoms (GSRS) and having comorbidities were associated with lower VAS and TTO or higher WTP values for current own health (p < 0.05). CONCLUSION This is the first study to report TTO utilities for CD health states. Strict adherence to the GFD may result in substantial health gains in symptomatic patients. Utilities and WTP results can be used to estimate benefits of GFD in cost-utility and cost-benefit analyses.
Collapse
Affiliation(s)
- M Mercédesz Angyal
- Károly Rácz Doctoral School of Clinical Medicine, Semmelweis University, 26 Üllői út, Budapest, 1085, Hungary
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary
| | - Peter L Lakatos
- McGill University Health Centre, Montreal General Hospital, 1650 Ave. Cedar, D16.173.1, Montreal, QC, H3G 1A4, Canada
- Department of Internal Medicine and Oncology, Semmelweis University, Korányi Sándor u. 2/a, Budapest, 1083, Hungary
| | - Balázs Jenei
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary
| | - Valentin Brodszky
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary
| | - Fanni Rencz
- Department of Health Policy, Corvinus University of Budapest, 8 Fővám tér, Budapest, 1093, Hungary.
| |
Collapse
|
38
|
King JA, Bakal JA, Li B, Whitten TA, Gidrewicz D, Turner JM, van Zanten SV, Huynh HQ, Heitman SJ, Shaheen AA, Quan H, Godley J, Underwood FE, Hracs L, Bergman D, Ludvigsson JF, Lebwohl B, Benchimol EI, Williamson T, Kaplan GG. Variation in Testing for and Incidence of Celiac Autoimmunity in Canada: A Population-Based Study. Gastroenterology 2023; 164:567-578.e7. [PMID: 36634826 DOI: 10.1053/j.gastro.2022.12.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS The incidence of biopsy-confirmed celiac disease has increased. However, few studies have explored the incidence of celiac autoimmunity based on positive serology results. METHODS A population-based cohort study assessed testing of tissue transglutaminase antibodies (tTG-IgA) in Alberta from 2012 to 2020. After excluding prevalent cases, incident celiac autoimmunity was defined as the first positive tTG-IgA result between 2015 and 2020. Testing and incidence rates for celiac autoimmunity were calculated per 1000 and 100,000 person-years, respectively. Incidence rate ratios (IRRs) were calculated to identify differences by demographic and regional factors. Average annual percent changes (AAPCs) assessed trends over time. RESULTS The testing rate of tTG-IgA was 20.2 per 1000 person-years and remained stable from 2012 to 2020 (AAPC, 1.2%; 95% confidence interval [CI], -0.5 to 2.9). Testing was higher in female patients (IRR, 1.66; 95% CI, 1.65-1.66), those living in metropolitan areas (IRR, 1.39; 95% CI, 1.38-1.40), and in areas of lower socioeconomic deprivation (lowest compared to highest IRR, 1.24; 95% CI, 1.23-1.25). Incidence of celiac autoimmunity was 33.8 per 100,000 person-years and increased from 2015 to 2020 (AAPC, 6.2%; 95% CI, 3.1-9.5). Among those with tTG-IgA results ≥10 times the upper limit of normal, the incidence was 12.9 per 100,000 person-years. The incidence of celiac autoimmunity was higher in metropolitan settings (IRR, 1.28; 95% CI, 1.21-1.35) and in the least socioeconomically deprived areas compared to the highest (IRR, 1.22; 95% CI, 1.14-1.32). CONCLUSIONS Incidence of celiac autoimmunity is high and increasing, despite stable testing rates. Variation in testing patterns may lead to underreporting the incidence of celiac autoimmunity in nonmetropolitan areas and more socioeconomically deprived neighborhoods.
Collapse
Affiliation(s)
- James A King
- Alberta Strategy for Patient Oriented Research Support Unit Data Platform, Calgary and Edmonton, Alberta, Canada; Provincial Research Data Services, Alberta Health Services, Calgary and Edmonton, Alberta, Canada; Centre for Health Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Jeffrey A Bakal
- Alberta Strategy for Patient Oriented Research Support Unit Data Platform, Calgary and Edmonton, Alberta, Canada; Provincial Research Data Services, Alberta Health Services, Calgary and Edmonton, Alberta, Canada; Department of General Internal Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Bing Li
- Alberta Strategy for Patient Oriented Research Support Unit Data Platform, Calgary and Edmonton, Alberta, Canada; Provincial Research Data Services, Alberta Health Services, Calgary and Edmonton, Alberta, Canada; Centre for Health Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Tara A Whitten
- Alberta Strategy for Patient Oriented Research Support Unit Data Platform, Calgary and Edmonton, Alberta, Canada; Provincial Research Data Services, Alberta Health Services, Calgary and Edmonton, Alberta, Canada; Centre for Health Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Dominica Gidrewicz
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | | - Hien Q Huynh
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Steven J Heitman
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Abdel Aziz Shaheen
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hude Quan
- Centre for Health Informatics, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Jenny Godley
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Sociology, University of Calgary, Calgary, Alberta, Canada
| | - Fox E Underwood
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Lindsay Hracs
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - David Bergman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden; Brommaplan Primary Health Care Center, Stockholm, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden; Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
| | - Benjamin Lebwohl
- Department of Medicine, Celiac Disease Center at Columbia University Medical Center, New York, New York
| | - Eric I Benchimol
- SickKids Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, Child Health Evaluative Sciences, SickKids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pediatrics and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Tyler Williamson
- Centre for Health Informatics, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Gilaad G Kaplan
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
39
|
Galipeau HJ, Verdu EF. Reset after RESET: insights from a negative trial in coeliac disease. Lancet Gastroenterol Hepatol 2023; 8:395-396. [PMID: 36898392 DOI: 10.1016/s2468-1253(23)00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 03/09/2023]
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
40
|
Chen ZE, Lee HE, Wu TT. Histologic evaluation in the diagnosis and management of celiac disease: practical challenges, current best practice recommendations and beyond. Hum Pathol 2023; 132:20-30. [PMID: 35932826 DOI: 10.1016/j.humpath.2022.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Celiac disease (CD) is an immunoallergic enteropathy affecting genetically susceptible individuals upon dietary exposure to gluten. In current clinical practice, the diagnosis of CD is based on a combination of clinical, serologic, and histologic factors with the possible exception of pediatric patients. Histopathologic evaluation of small intestinal tissue plays a critical role in the disease diagnosis and management, despite many practical challenges. Recently published best practice guidelines help to standardize biopsy sample procurement, tissue preparation, histology interpretation, and reporting, to optimize patient care. In addition, an increasing demand for monitoring the disease course, particularly demonstrating the efficacy of dietary and nondietary interventions for disease management, calls for the use of quantitative histology. With the advent of a gradual transition toward digital pathology in routine diagnostic practice, quantitative histopathologic evaluation in CD shows a promising future.
Collapse
Affiliation(s)
- Zongming Eric Chen
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hee Eun Lee
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tsung-Teh Wu
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
41
|
Mamone G, Di Stasio L, Vitale S, Picascia S, Gianfrani C. Analytical and functional approaches to assess the immunogenicity of gluten proteins. Front Nutr 2023; 9:1049623. [PMID: 36741992 PMCID: PMC9890883 DOI: 10.3389/fnut.2022.1049623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/15/2022] [Indexed: 01/19/2023] Open
Abstract
Gluten proteins are the causative agents of celiac disease (CD), a lifelong and worldwide spread food intolerance, characterized by an autoimmune enteropathy. Gluten is a complex mixture of high homologous water-insoluble proteins, characterized by a high content of glutamine and proline amino acids that confers a marked resistance to degradation by gastrointestinal proteases. As a consequence of that, large peptides are released in the gut lumen with the potential to activate inflammatory T cells, in CD predisposed individuals. To date, several strategies aimed to detoxify gluten proteins or to develop immunomodulatory drugs to recover immune tolerance to gluten are under investigation. This review overviews the state of art of both analytical and functional methods currently used to assess the immunogenicity potential of gluten proteins from different cereal sources, including native raw seed flours and complex food products, as well as drug-treated samples. The analytical design to assess the content and profile of gluten immunogenic peptides, described herein, is based on the oral-gastro-intestinal digestion (INFOGEST model) followed by extensive characterization of residual gluten peptides by proteomic and immunochemical analyses. These approaches include liquid chromatography-high-resolution mass spectrometry (LC-MS/MS) and R5/G12 competitive ELISA. Functional studies to assess the immune stimulatory capabilities of digested gluten peptides are based on gut mucosa T cells or peripheral blood cells obtained from CD volunteers after a short oral gluten challenge.
Collapse
Affiliation(s)
- Gianfranco Mamone
- Institute of Food Science, Department of Biology, Agriculture and Food Sciences, National Research Council of Italy, Avellino, Italy
| | - Luigia Di Stasio
- Institute of Food Science, Department of Biology, Agriculture and Food Sciences, National Research Council of Italy, Avellino, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy
| | - Stefania Picascia
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy,*Correspondence: Carmen Gianfrani,
| |
Collapse
|
42
|
Gromny I, Neubauer K. Pancreatic Cancer in Celiac Disease Patients-A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1565. [PMID: 36674320 PMCID: PMC9867303 DOI: 10.3390/ijerph20021565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 06/01/2023]
Abstract
Background: Celiac disease (CD) is an autoimmune enteropathy affecting approximately 1% of the population and is associated with an increased risk of enteropathy-associated T-cell lymphoma and small bowel adenocarcinoma, whereas the association between CD and other malignancies is unclear. Since pancreatic cancer (PC) remains one of the most lethal neoplasms and its incidence is increasing despite numerous ongoing research on diagnostic biomarkers and novel therapies, we aimed to investigate whether CD has an impact on the risk of PC. Material and Methods: We performed a systematic review of the literature published from January 2000 to March 2022 in two databases: Web of Science and Scopus and a meta-analysis of eligible studies. Results: Our search identified eight publications included in the systematic review. A total of five studies involving 47,941 patients, including 6399 CD patients with malignancies and 1231 PC cases were included in the meta-analysis and 221 cases of PC in CD patients with other cancers were recognized. The pooled OR for PC was 1.46 (95% CI 1.26−1.7) with significant heterogeneity (89.1%; p < 0.05), suggesting that CD patients with malignancies were at higher risk for PC. Conclusions: The association between CD and PC is uncertain. However, the results of the current meta-analysis may indicate an increased risk of PC in the group of patients with CD and other cancers. Further multicenter studies are warranted.
Collapse
Affiliation(s)
- Iga Gromny
- Division of Dietetics, Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Katarzyna Neubauer
- Division of Dietetics, Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| |
Collapse
|
43
|
Yuan S, Kim JH, Xu P, Wang Z. Causal association between celiac disease and inflammatory bowel disease: A two-sample bidirectional Mendelian randomization study. Front Immunol 2023; 13:1057253. [PMID: 36685511 PMCID: PMC9845610 DOI: 10.3389/fimmu.2022.1057253] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Background An epidemiological link between celiac disease (CeD) and inflammatory bowel disease (IBD) has been well established recently. In this study, Mendelian randomization (MR) analysis was performed employing pooled data of publicly available genome-wide association studies (GWAS) to determine the causal relationship between CeD and IBD, encompassing ulcerative colitis (UC) and Crohn's disease (CD). Methods Dataset of CeD was acquired from GWAS for 12,041 cases and 12,228 controls. A GWAS of more than 86,000 patients and controls was used to identify genetic variations underlying IBD. MR analyses were performed with an inverse-variance-weighted approach, an MR-Egger regression, a weighted-mode approach, a weighted-median method, and sensitivity analyses of MR pleiotropy residual sum and outlie (MR-PRESSO). Results MR demonstrated that genetic predisposition to CeD was linked to a augmented risk of IBD (OR: 1.1408; 95% CI: 1.0614-1.2261; P = 0.0003). In the analysis of the two IBD subtypes, genetic predisposition to CeD was also linked to increased risks of UC (OR: 1.1646; 95% CI: 1.0614-1.2779; P = 0.0012) and CD (OR: 1.1865; 95% CI: 1.0948-1.2859; P = 3.07E-05). Reverse MR analysis results revealed that genetic susceptibility to IBD and CD was correlated with an augmented risk of CeD. However, there was no genetic correlation between UC and CeD. All of the above results were validated with other GWAS databases. Conclusion There is a bidirectional causal relationship of CeD with IBD and CD. However, UC only augments the risk of developing CeD.
Collapse
Affiliation(s)
- Shuai Yuan
- Division of Pancreatobiliary Surgery, Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ji Hun Kim
- Division of Pancreatobiliary Surgery, Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea,*Correspondence: Ji Hun Kim,
| | - Pai Xu
- Department of Orthopaedic Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Zhao Wang
- Department of Orthopaedic Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
44
|
Machado MV. New Developments in Celiac Disease Treatment. Int J Mol Sci 2023; 24:ijms24020945. [PMID: 36674460 PMCID: PMC9862998 DOI: 10.3390/ijms24020945] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/18/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Celiac disease (CD) is a common autoimmune disease affecting around 1% of the population. It consists of an immune-mediated enteropathy, triggered by gluten exposure in susceptible patients. All patients with CD, irrespective of the presence of symptoms, must endure a lifelong gluten-free diet (GFD). This is not an easy task due to a lack of awareness of the gluten content in foods and the extensive incorporation of gluten in processed foods. Furthermore, a GFD imposes a sense of limitation and might be associated with decreased quality of life in CD patients. This results in gluten contamination in the diet of four out of five celiac patients adhering to a GFD. Furthermore, one in three adult patients will report persistent symptoms and two in three will not achieve full histological recovery when on a GFD. In recent years, there has been extensive research conducted in the quest to find the holy grail of pharmacological treatment for CD. This review will present a concise description of the current rationale and main clinical trials related to CD drug therapy.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Gastroenterology Department, Hospital de Vila Franca de Xira, Estrada Carlos Lima Costa, Nª 2, 2600-009 Vila Franca de Xira, Portugal; ; Tel.: +351-263-006-500
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
45
|
Dewala S, Bodkhe R, Nimonkar Y, Prakash OM, Ahuja V, Makharia GK, Shouche YS. Human small-intestinal gluten-degrading bacteria and its potential implication in celiac disease. J Biosci 2023; 48:18. [PMID: 37309172 DOI: 10.1007/s12038-023-00337-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/11/2023] [Indexed: 08/30/2023]
Abstract
Celiac disease (CeD) is an immune-mediated chronic disorder triggered by the ingestion of wheat gluten in genetically predisposed individuals. Gluten is a major food ingredient, infamously containing proline and glutamine-rich domains that are highly resistant to digestion by mammalian proteolytic enzymes. Thus, adhering to a gluten-free diet (GFD) is the only known treatment for CeD, albeit with many complications. Therefore, any therapy that eliminates the gluten immunogenic part before it reaches the small intestine is highly desirable. Probiotic therapy containing gluten-degrading bacteria (GDB) and their protease enzymes are possibly new approaches to treating CeD. Our study aimed to identify novel GDB from the duodenal biopsy of the first-degree relative (FDR) subjects (relatives of diseased individuals who are healthy but susceptible to celiac disease) with the potential to reduce gluten immunogenicity. Using the gluten agar plate technique, bacterial strains Brevibacterium casei NAB46 and Staphylococcus arlettae R2AA77 displaying glutenase activity were screened, identified, and characterized. Whole-genome sequencing found gluten-degrading prolyl endopeptidase (PEP) in the B. casei NAB46 genome and glutamyl endopeptidase (GEP) in the S. arlettae R2AA77 genome. Partially purified PEP has a specific activity of 1.15 U/mg, while GEP has a specific activity of 0.84 U/mg, which are, respectively, 6- and 9-fold times higher after concentrating the enzymes. Our results showed that these enzymes could hydrolyse immunotoxic gliadin peptides recognized in western blot using an anti-gliadin antibody. Additionally, a docking model was proposed for representative gliadin peptide PQPQLPYPQPQLP in the active site of the enzymes, where the residues of the N-terminal peptide extensively interact with the catalytic domain of the enzymes. These bacteria and their associated glutenase enzymes efficiently neutralize gliadin immunogenic epitopes, opening possibilities for their application as a dietary supplement in treating CeD patients.
Collapse
|
46
|
Abstract
Proteases are an evolutionarily conserved family of enzymes that degrade peptide bonds and have been implicated in several common gastrointestinal (GI) diseases. Although luminal proteolytic activity is important for maintenance of homeostasis and health, the current review describes recent advances in our understanding of how overactivity of luminal proteases contributes to the pathophysiology of celiac disease, irritable bowel syndrome, inflammatory bowel disease and GI infections. Luminal proteases, many of which are produced by the microbiota, can modulate the immunogenicity of dietary antigens, reduce mucosal barrier function and activate pro-inflammatory and pro-nociceptive host signaling. Increased proteolytic activity has been ascribed to both increases in protease production and decreases in inhibitors of luminal proteases. With the identification of strains of bacteria that are important sources of proteases and their inhibitors, the stage is set to develop drug or microbial therapies to restore protease balance and alleviate disease.
Collapse
Affiliation(s)
- Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mabel Guzman
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada
| | - Josie Libertucci
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Alan E. Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada,CONTACT Alan E. Lomax Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
47
|
Ruigrok RAAA, Weersma RK, Vich Vila A. The emerging role of the small intestinal microbiota in human health and disease. Gut Microbes 2023; 15:2201155. [PMID: 37074215 PMCID: PMC10120449 DOI: 10.1080/19490976.2023.2201155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
The human gut microbiota continues to demonstrate its importance in human health and disease, largely owing to the countless number of studies investigating the fecal microbiota. Underrepresented in these studies, however, is the role played by microbial communities found in the small intestine, which, given the essential function of the small intestine in nutrient absorption, host metabolism, and immunity, is likely highly relevant. This review provides an overview of the methods used to study the microbiota composition and dynamics along different sections of the small intestine. Furthermore, it explores the role of the microbiota in facilitating the small intestine in its physiological functions and discusses how disruption of the microbial equilibrium can influence disease development. The evidence suggests that the small intestinal microbiota is an important regulator of human health and its characterization has the potential to greatly advance gut microbiome research and the development of novel disease diagnostics and therapeutics.
Collapse
Affiliation(s)
- Renate A. A. A. Ruigrok
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
48
|
Furfaro F, Gabbiadini R, D'Amico F, Zilli A, Dal Buono A, Allocca M, Fiorino G, Danese S. Gastrointestinal System: COVID-19 and Potential Mechanisms Associated with Coagulopathy. Curr Drug Targets 2022; 23:1611-1619. [PMID: 36154571 DOI: 10.2174/1389450123666220922095913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/25/2023]
Abstract
SARS-CoV-2 is a novel coronavirus that expanded worldwide, generating a pandemic of acute respiratory syndrome called "coronavirus disease 2019" (COVID-19), which resulted in a global health crisis. The spectrum of COVID-19 manifestations ranges from none or mild symptoms to severe respiratory failure associated with systemic manifestations, mostly gastrointestinal symptoms. Hypercoagulability is an important feature of COVID-19 disease, which can potentially influence patients' prognosis. Therefore, gastroenterologists should focus on subjects with concomitant hypercoagulable gastrointestinal disorders as they may display a higher risk of thrombotic complications during SARS-CoV-2 infection. The aim of this review is to summarize the available evidence regarding the interplay of the prothrombotic pathogenetic mechanisms of both COVID-19 and hypercoagulable digestive diseases and the possible clinical implications. We summarized the potential interplay of prothrombotic mechanisms of both COVID-19 and hypercoagulable digestive diseases in the graphical abstract.
Collapse
Affiliation(s)
- Federica Furfaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ferdinando D'Amico
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Alessandra Zilli
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mariangela Allocca
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Gionata Fiorino
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
49
|
Virta J, Hannula M, Lindfors K, Tamminen I, Taavela J, Huhtala H, Kaukinen K, Saavalainen P, Hyttinen J, Kurppa K. Validation of the X-ray microtomography in the assessment of duodenal morphometry and surface area in celiac disease. Front Immunol 2022; 13:945197. [PMID: 36211435 PMCID: PMC9539806 DOI: 10.3389/fimmu.2022.945197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Duodenal histology remains the diagnostic reference standard in celiac disease. However, traditional methods have suboptimal sensitivity and reproducibility for early mucosal changes and research purposes. We validated a recently introduced micro-CT imaging method for an accurate digital evaluation of duodenal histomorphometry and mucosal surface areas. Methods Endoscopic biopsies from 58 individuals were utilized for the micro-CT imaging, selecting histological changes ranging from normal to severely damaged mucosa. The imaging protocol was optimized for practicability and resolution. The Bland–Altman method was applied to test intra- and interobserver variations in the blinded measurements. Results The 3D micro-CT reconstructions enabled easy and precise digital cutting with optimal orientation and computer-assisted measurement of the surface area. Intraobserver analysis of morphological measurements showed a mean difference of 0.011 with limits of agreement (LA) from -0.397 to 0.375 and a standard deviation (SD) of 0.197. The corresponding figures for interobserver analysis were 0.080, from -0.719 to 0.537 and 0.320, respectively. The intraclass correlation coefficients (ICC) for the intraobserver and interobserver variations were 0.981 and 0.954, respectively. Intraobserver surface area analysis yielded a mean difference of 0.010, LA from -0.764 to 0.785 and an SD of 0.395, and an interobserver analysis mean difference of 0.028, LA from -0.642 to 0.698 and SD of 0.342. The respective ICCs for the intra- and interobserver variations were 0.963 and 0.972. Conclusions Micro-CT showed excellent accuracy and reproducibility in the evaluation of mucosal morphometry and surface areas. The improved sensitivity for histological changes is a powerful tool for the diagnosis of celiac disease and for clinical and pharmacological studies.
Collapse
Affiliation(s)
- Johannes Virta
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Markus Hannula
- Computational Biophysics and Imaging Group, The Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ilmari Tamminen
- Computational Biophysics and Imaging Group, The Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juha Taavela
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Katri Kaukinen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Päivi Saavalainen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, The Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kalle Kurppa
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- The University Consortium of Seinäjoki and Seinäjoki Central Hospital, Seinäjoki, Finland
- *Correspondence: Kalle Kurppa,
| |
Collapse
|
50
|
Chan AP, Jun-Ihn E, Shrestha G, Rao L, Vahabnezhad E, Vargas J. A 15-Month-Old With Developmental Regression and Hypotonia. Clin Pediatr (Phila) 2022; 62:368-371. [PMID: 36146920 DOI: 10.1177/00099228221124678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Alvin P Chan
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Esther Jun-Ihn
- Division of Pediatric Hospital Medicine, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Gyaneshwar Shrestha
- Division of Pediatric Neurology, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lekha Rao
- Division of Pediatric Neurology, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Elaheh Vahabnezhad
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jorge Vargas
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, UCLA Mattel Children's Hospital and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|