1
|
Hracs L, Windsor JW, Gorospe J, Cummings M, Coward S, Buie MJ, Quan J, Goddard Q, Caplan L, Markovinović A, Williamson T, Abbey Y, Abdullah M, Abreu MT, Ahuja V, Raja Ali RA, Altuwaijri M, Balderramo D, Banerjee R, Benchimol EI, Bernstein CN, Brunet-Mas E, Burisch J, Chong VH, Dotan I, Dutta U, El Ouali S, Forbes A, Forss A, Gearry R, Dao VH, Hartono JL, Hilmi I, Hodges P, Jones GR, Juliao-Baños F, Kaibullayeva J, Kelly P, Kobayashi T, Kotze PG, Lakatos PL, Lees CW, Limsrivilai J, Lo B, Loftus EV, Ludvigsson JF, Mak JWY, Miao Y, Ng KK, Okabayashi S, Olén O, Panaccione R, Paudel MS, Quaresma AB, Rubin DT, Simadibrata M, Sun Y, Suzuki H, Toro M, Turner D, Iade B, Wei SC, Yamamoto-Furusho JK, Yang SK, Ng SC, Kaplan GG. Global evolution of inflammatory bowel disease across epidemiologic stages. Nature 2025:10.1038/s41586-025-08940-0. [PMID: 40307548 DOI: 10.1038/s41586-025-08940-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
During the twentieth century, inflammatory bowel disease (IBD) was considered a disease of early industrialized regions in North America, Europe and Oceania1. At the turn of the twenty-first century, IBD incidence increased in newly industrialized and emerging regions in Africa, Asia and Latin America, while the prevalence in early industrialized regions continued to grow steadily2-4. Changes in the incidence and prevalence denote the evolution of IBD across four epidemiologic stages: stage 1 (emergence), characterized by low incidence and prevalence; stage 2 (acceleration in incidence), marked by rapidly rising incidence and low prevalence; and stage 3 (compounding prevalence), where the incidence decelerates, plateaus or declines while the prevalence steadily increases. A fourth stage (prevalence equilibrium) has been proposed in which the prevalence slope plateaus due to demographic shifts in an ageing IBD population, but it has not yet been evidenced. To date, these stages have remained theoretical, lacking specific numerical indicators to define transition points. Here, using real-world data from 522 population-based studies encompassing 82 global regions and spanning more than a century (1920-2024), we show spatiotemporal transitions across stages 1-3 and model stage 4 progression. Understanding the evolution of IBD across epidemiologic stages enables healthcare systems to better anticipate the future worldwide burden of IBD.
Collapse
Affiliation(s)
- Lindsay Hracs
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joseph W Windsor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Julia Gorospe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael Cummings
- Centre for Health Informatics and Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie Coward
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael J Buie
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joshua Quan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Quinn Goddard
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Léa Caplan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ante Markovinović
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tyler Williamson
- Centre for Health Informatics and Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Yvonne Abbey
- Maidstone and Tunbridge Wells NHS Trust, Kent, UK
| | - Murdani Abdullah
- Division of Gastroenterology, Department of Internal Medicine, HCRC IMERI, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Maria T Abreu
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
| | - Vineet Ahuja
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Raja Affendi Raja Ali
- Sir Jeffrey Cheah Sunway Medical School, Faculty of Medical and Life Sciences, Sunway University, Selangor, Malaysia
| | - Mansour Altuwaijri
- Division of Gastroenterology, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Domingo Balderramo
- IBD Unit, Gastroenterology Department, Hospital Privado Universitario de Córdoba, Instituto Universitario de Ciencias Biomédicas de Córdoba, Córdoba, Argentina
| | - Rupa Banerjee
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- IBD Center, Asian Institute of Gastroenterology, Hyderabad, India
| | - Eric I Benchimol
- SickKids Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children (SickKids), Toronto, Ontario, Canada
- Department of Paediatrics and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Charles N Bernstein
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Department of Medicine, and the University of Manitoba IBD Clinical and Research Centre, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eduard Brunet-Mas
- Gastroenterology Department, Parc Taulí Hospital Universitari, Institut d'Investigació I Innovació Parc Taulí (I3PT-CERCA), Sabadell, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Johan Burisch
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Gastrounit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vui Heng Chong
- Division of Gastroenterology and Hepatology, Department of Medicine, Raja Isteri Pengiran Anak Saleha (RIPAS) Hospital, Bandar Seri Begawan, Brunei Darussalam
| | - Iris Dotan
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Usha Dutta
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sara El Ouali
- Digestive Disease Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
- Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Angela Forbes
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Anders Forss
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Richard Gearry
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Viet Hang Dao
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Juanda Leo Hartono
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division Gastroenterology & Hepatology, National University Hospital, Singapore, Singapore
| | - Ida Hilmi
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Phoebe Hodges
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, UK
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Gareth-Rhys Jones
- University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| | - Fabián Juliao-Baños
- Department of Gastroenterology, Pablo Tobon Uribe Hospital, Medellín, Colombia
| | - Jamilya Kaibullayeva
- JSC Research Institute of Cardiology and Internal Diseases of the Ministry of Health of the Republic of Kazakhstan, Astana, Republic of Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Paul Kelly
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, UK
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Paulo Gustavo Kotze
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Health Sciences Postgraduate Program, Pontificia Universidade Católica do Paraná, Curitiba, Brazil
| | - Peter L Lakatos
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Division of Gastroenterology, McGill University, Montreal, Quebec, Canada
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Charlie W Lees
- Centre for Genomics and Experimental Medicine (CGEM), Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh IBD Unit, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Julajak Limsrivilai
- Division of Gastroenterology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Bobby Lo
- Gastrounit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Edward V Loftus
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Örebro University Hospital, Stockholm, Sweden
| | - Joyce W Y Mak
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - YingLei Miao
- Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Ka Kei Ng
- Conde S. Januário Hospital, Macao SAR, China
| | - Shinji Okabayashi
- Department of Healthcare Epidemiology, School of Public Health in the Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ola Olén
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Remo Panaccione
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
| | - Mukesh Sharma Paudel
- Department of Gastroenterology, National Academy of Medical Sciences, Kathmandu, Nepal
| | - Abel Botelho Quaresma
- UNOESC Curso de Medicina: Universidade do Oeste de Santa Catarina, Joaçaba, Brazil
- Health Sciences Postgraduate Program, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - David T Rubin
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- Inflammatory Bowel Disease Center, University of Chicago Medicine, Chicago, IL, USA
| | - Marcellus Simadibrata
- Division of Gastroenterology, Department of Internal Medicine, HCRC IMERI, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Yang Sun
- Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Hidekazu Suzuki
- Division of Gastroenteroloy and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Martin Toro
- Head of the Inflammatory Bowel Diseases Unit, HIGEA, Mendoza, Argentina
| | - Dan Turner
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden
- The Juliet Keidan Institute of Pediatric Gastroenterology & Nutrition, The Hebrew University of Jerusalem, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Beatriz Iade
- Cooperativa de Servicios Médicos (COSEM), Montevideo, Uruguay
| | - Shu Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Jesus K Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, National Institute of Medical Sciences and Nutrition and National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Siew C Ng
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden.
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Microbiota I-Center (MagIC), Hong Kong, China.
- New Cornerstone Science Laboratory, The Chinese University of Hong Kong, Hong Kong, China.
| | - Gilaad G Kaplan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada.
- International Organization for the study of Inflammatory Bowel Disease (IOIBD), Stockholm, Sweden.
| |
Collapse
|
2
|
Ng WK, Thanusha G, Chong PP, Chuah C. The Power of Antibodies: Advancing Biomarker-Based Disease Detection and Surveillance. Immunol Invest 2025:1-25. [PMID: 40256875 DOI: 10.1080/08820139.2025.2492246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
BACKGROUND Antibodies have long served as fundamental tools in disease diagnosis and surveillance. Their utility as biomarkers has expanded beyond infectious diseases to encompass a wide range of health conditions. OBJECTIVES This review aims to explore recent advancements in antibody biomarker discovery and their applications in diagnosing and monitoring diverse health conditions. It also examines the role of antibody surveillance in public health and epidemiological studies. METHODS A comprehensive analysis of recent literature was conducted, focusing on studies that identify and characterize disease-specific antibodies. Particular attention was given to their relevance in autoimmune diseases, infections, cancers, and neurological disorders. CONTENT The review highlights disease-specific antibody biomarkers and their clinical significance. It also discusses the utility and challenges of antibody-based surveillance in assessing disease prevalence, tracking immunity trends, and supporting One Health strategies. CONCLUSIONS Recent advancements in antibody biomarker discovery demonstrate significant potential in improving early diagnosis, personalized treatment, and population-level health management. Antibody surveillance continues to play a pivotal role in guiding public health responses and understanding disease dynamics.
Collapse
Affiliation(s)
- Woei Kean Ng
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| | - Gunasegran Thanusha
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Candy Chuah
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| |
Collapse
|
3
|
Taylor H, Uhlig HH, Powrie F. Autoimmunity in inflammatory bowel disease: a holobiont perspective. Curr Opin Immunol 2025; 94:102557. [PMID: 40252635 DOI: 10.1016/j.coi.2025.102557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/21/2025]
Abstract
Adaptive immunity towards self-antigens (autoimmunity) and intestinal commensal microbiota is a key feature of inflammatory bowel disease (IBD). Considering mucosal adaptive immunity from a holobiont perspective, where the host and its microbiome form a single physiological unit, emphasises the challenge of avoiding damaging responses to self-antigen and symbiotic microbial communities in the gut while protecting against potential pathogens. Intestinal tolerance mechanisms prevent maladaptive T and B cell responses to microbial, environmental, and self-antigens, which drive inflammation. We discuss the spectrum of antimicrobial and autoantibody responses and highlight mechanisms by which common IBD-associated adaptive immune responses contribute to disease.
Collapse
Affiliation(s)
- Henry Taylor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Holm H Uhlig
- Centre for Human Genetics, University of Oxford, Oxford, UK; Translational Gastroenterology Liver Unit, University of Oxford, Oxford, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Chhibba T, Gros B, King JA, Windsor JW, Gorospe J, Leibovitzh H, Xue M, Turpin W, Croitoru K, Ananthakrishnan AN, Gearry RB, Kaplan GG. Environmental risk factors of inflammatory bowel disease: toward a strategy of preventative health. J Crohns Colitis 2025; 19:jjaf042. [PMID: 40065502 PMCID: PMC12010164 DOI: 10.1093/ecco-jcc/jjaf042] [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] [Indexed: 04/22/2025]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) involves a complex interplay between genetic, environmental, and microbial factors. Many of these environmental determinants are modifiable, offering opportunities to prevent disease or delay its onset. Advances in the study of preclinical IBD cohorts offer the potential to identify biomarkers that predict individuals at high risk of developing IBD, enabling targeted environmental interventions aimed at reducing IBD incidence. This review summarizes findings from 79 meta-analyses on modifiable environmental factors associated with the development of IBD. Identified risk factors include smoking, Western diets, ultra-processed foods, and early life antibiotic use, while protective factors include breastfeeding, Mediterranean diets rich in fiber, plant-based foods, and fish, along with an active physical lifestyle. Despite the promise shown by observational data, interventional or randomized controlled studies evaluating the efficacy of modifying environmental risk factors remain limited and mostly focus on dietary intervention. This review aims to inform the design of higher quality interventional and randomized controlled studies for disease prevention while providing actionable guidance to healthcare providers on reducing the risk of developing IBD through environmental modifications.
Collapse
Affiliation(s)
- Tarun Chhibba
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Beatriz Gros
- Department of Gastroenterology and Hepatology, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Madrid, Spain
| | - James A King
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Joseph W Windsor
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Julia Gorospe
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Haim Leibovitzh
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mingyue Xue
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Williams Turpin
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Richard B Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Gilaad G Kaplan
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Grännö O, Bergemalm D, Salomon B, Lindqvist CM, Hedin CRH, Carlson M, Dannenberg K, Andersson E, Keita ÅV, Magnusson MK, Eriksson C, Lanka V, Magnusson PKE, D'Amato M, Öhman L, Söderholm JD, Hultdin J, Kruse R, Cao Y, Repsilber D, Grip O, Karling P, Halfvarson J. Preclinical Protein Signatures of Crohn's Disease and Ulcerative Colitis: A Nested Case-Control Study Within Large Population-Based Cohorts. Gastroenterology 2025; 168:741-753. [PMID: 39608683 DOI: 10.1053/j.gastro.2024.11.006] [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: 01/17/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND & AIMS Biomarkers are needed to identify individuals at elevated risk of inflammatory bowel disease. This study aimed to identify protein signatures predictive of inflammatory bowel disease. METHODS Using large population-based cohorts (n ≥180,000), blood samples were obtained from individuals who later in life were diagnosed with inflammatory bowel disease and compared with age and sex-matched controls, free from inflammatory bowel disease during follow-up. A total of 178 proteins were measured on Olink platforms. We used machine-learning methods to identify protein signatures of preclinical disease in the discovery cohort (n = 312). Their performance was validated in an external preclinical cohort (n = 222) and assessed in an inception cohort (n = 144) and a preclinical twin cohort (n = 102). RESULTS In the discovery cohort, a signature of 29 proteins differentiated preclinical Crohn's disease (CD) cases from controls, with an area under the curve (AUC) of 0.85. Its performance was confirmed in the preclinical validation (AUC = 0.87) and the inception cohort (AUC = 1.0). In preclinical samples, downregulated (but not upregulated) proteins related to gut barrier integrity and macrophage functionality correlated with time to diagnosis of CD. The preclinical ulcerative colitis signature had a significant, albeit lower, predictive ability in the discovery (AUC = 0.77), validation (AUC = 0.67), and inception cohorts (AUC = 0.95). The preclinical signature for CD demonstrated an AUC of 0.89 when comparing twins with preclinical CD with matched external healthy twins, but its predictive ability was lower (AUC = 0.58; P = .04) when comparing them with their healthy twin siblings, that is, when accounting for genetic and shared environmental factors. CONCLUSION We identified protein signatures for predicting a future diagnosis of CD and ulcerative colitis, validated across independent cohorts. In the context of CD, the signature offers potential for early prediction.
Collapse
Affiliation(s)
- Olle Grännö
- Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Daniel Bergemalm
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Benita Salomon
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Carl Mårten Lindqvist
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Charlotte R H Hedin
- Gastroenterology Unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Marie Carlson
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Katharina Dannenberg
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Erik Andersson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria K Magnusson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Carl Eriksson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden; Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vivekananda Lanka
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mauro D'Amato
- Department of Medicine and Surgery, LUM University, Casamassima, Italy; Gastrointestinal Genetics Lab, CIC BioGUNE-BRTA, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Johan D Söderholm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Surgery, Linköping University, Linköping, Sweden
| | - Johan Hultdin
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Robert Kruse
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden; Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Dirk Repsilber
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Olof Grip
- Department of Gastroenterology, Skåne University Hospital, Malmö, Sweden
| | - Pontus Karling
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
6
|
Nakanishi R, Kuwada T, Shiokawa M, Nishikawa Y, Ota S, Yamazaki H, Yanaidani T, Sawada K, Hirata A, Yasuda M, Takimoto I, Chikugo K, Yokode M, Muramoto Y, Matsumoto S, Matsumori T, Uza N, Chiba T, Seno H. Anti-integrin αvβ6 Antibodies Predict Pouchitis in Patients With Ulcerative Colitis After Restorative Proctocolectomy With Ileal Pouch-Anal Anastomosis. Inflamm Bowel Dis 2025; 31:777-785. [PMID: 39657162 PMCID: PMC11890122 DOI: 10.1093/ibd/izae263] [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: 07/25/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Pouchitis is the most common complication of restorative proctocolectomy (RPC) with ileal pouch-anal anastomosis (IPAA) in patients with ulcerative colitis (UC). We previously reported the presence of anti-integrin αvβ6 antibodies in the serum of patients with UC. This study investigated the association between anti-integrin αvβ6 antibodies and the development of pouchitis in patients with UC. METHODS Serum levels of anti-integrin αvβ6 antibodies were measured by enzyme-linked immunosorbent assay in 16 patients with UC who underwent RPC with IPAA. Integrin αvβ6 expression in the colonic, terminal ileal, and pouch epithelium was examined using immunohistochemistry and western blot analysis. RESULTS Anti-integrin αvβ6 antibody levels in patients with UC were significantly decreased at 3, 9, and 12 months after RPC (P < .05). However, in patients who developed pouchitis, antibody levels remained high. The antibody levels at the time of RPC were significantly higher in patients who developed pouchitis compared to those who did not. Kaplan-Meier analysis revealed a significantly higher incidence of pouchitis in patients with antibody levels above the cutoff at the time of RPC. Although integrin αvβ6 was not expressed in the terminal ileal epithelium at the time of RPC, expression became positive in the pouch epithelium of patients with pouchitis. CONCLUSIONS The anti-integrin αvβ6 antibody levels in patients with UC were decreased after RPC but remained high in patients who developed pouchitis. The antibody levels at the time of RPC may serve as a potential prognostic biomarker for predicting the risk of pouchitis in patients with UC.
Collapse
Affiliation(s)
- Risa Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshihiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sakiko Ota
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takafumi Yanaidani
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Sawada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ayako Hirata
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Muneji Yasuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuhisa Takimoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koki Chikugo
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Yokode
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuya Muramoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shimpei Matsumoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Kansai Electric Power Hospital, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
7
|
Venkat S, Rusbuldt J, Richards D, Freeman T, Richmond C, Mortensen JH, Seidelin JB, Poulsen A, McRae B, Ruane D. Serum Collagen Biomarkers Are Reflective of Tissue Specific Fibroblasts Associated With Ulcerative Colitis Activity and Treatment Response to Ustekinumab. United European Gastroenterol J 2025. [PMID: 39969502 DOI: 10.1002/ueg2.70002] [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: 10/25/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND There is a need to identify peripheral biomarkers reflective of defined disease associated fibroblasts in Ulcerative Colitis (UC), with the aim of enabling clinical development approaches for novel-stromal-targeted therapeutics for individuals at risk for fibrostenotic complications. Additionally, longitudinal non-invasive biomarkers of tissue remodelling, fibroblast biology and pharmacodynamic measurements are needed in the clinic to facilitate risk stratification. AIM To identify novel blood protein biomarkers associated with defined fibroblast subsets, tissue remodelling and treatment response/non-response in UC. METHODS We performed data analysis on matched serum and tissue transcriptomics from the UNIFI trial at weeks 0 and 8 in clinical responders and non-responders. Detailed gene correlation analysis was performed on 97 colonic biopsies from 50 patients pre- and post-treatment, to construct detailed cell-type mapping associated with clinical parameters. Detailed serum-based proteomics analysis was performed using matched serum and tissue sample sets to evaluate specific correlations between defined tissue cellular subsets and unique peripheral proteins, reflective of defined tissue transcriptional subsets and clinical parameters. RESULTS Evaluation of the UNIFI clinical study, revealed a significant association between intestinal-inflammatory activated fibroblasts (IIAF) and various clinical parameters, including Geboes scores. These findings were unique to IIAFs and were confirmed using spatial tissue transcriptomics. Evaluation of novel peripheral proteomics revealed a significant correlation between selective serum collagen biomarkers, including Pro-Collagen 22, Collagen 1M, CTX-III, ELP-3, and the IIAF tissue module. These serum collagen biomarkers were unique to IIAFs, as other broad proteomics methodologies failed to demonstrate significant correlations with known UC serum markers. Ustekinumab endoscopic responders had a significant decrease in IIAFs, which was associated with decreases in these IIAF associated serum proteins. Furthermore, C1M and ELP-3 demonstrated predictive value to enable characterisation of UC patients with IIAF driven disease. CONCLUSIONS These serum biomarkers were correlated with tissue levels of IIAFs, identifying unique peripheral markers of tissue associated cell types correlated with fibrosis. Given the association of IIAFs and treatment response, this highlights the utility of these triaged collagen biomarkers for anti-stromal therapeutic development and patient stratification in UC and beyond.
Collapse
Affiliation(s)
- Swati Venkat
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | - Joshua Rusbuldt
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | - Dylan Richards
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | - Thomas Freeman
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | - Camilla Richmond
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | | | - Jakob Benedict Seidelin
- Department of Digestive Diseases, Transplantation and General Surgery, Section for IBD, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Anja Poulsen
- Department of Digestive Diseases, Transplantation and General Surgery, Section for IBD, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Brad McRae
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| | - Darren Ruane
- Janssen Immunology, Translation Sciences & Medicine, Spring House, Pennsylvania, USA
| |
Collapse
|
8
|
Barnes EL. Onset of pouchitis after colectomy for ulcerative colitis: reducing the risk. Expert Rev Gastroenterol Hepatol 2025. [PMID: 39907581 DOI: 10.1080/17474124.2025.2464058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Pouchitis remains the most common complication after ileal pouch-anal anastomosis (IPAA) for ulcerative colitis (IPAA). Despite the burden of pouchitis, our ability to recognize the patients at greatest risk for pouchitis and to offer early intervention for the prevention or early treatment is limited. AREAS COVERED In this review, we will discuss the diagnostic categories of pouchitis, as well as the common approaches to treatment. We will then discuss gaps in research and clinical care. These include opportunities to improve our understanding of the risk factors associated with the development of pouchitis as well as the role that primary and secondary prevention may play in the management of patients after IPAA to prevent pouchitis. We will then discuss future research goals to decrease the burden of pouchitis. EXPERT OPINION Although the burden of pouchitis is well recognized, we have an opportunity to improve our methods of risk stratification to offer early intervention to those patients at the greatest risk for developing pouchitis. Understanding which preventive therapies, in which populations, and under which treatment settings offer the most benefit will be critical issues to address in the near future to improve care of patients after IPAA for UC.
Collapse
Affiliation(s)
- Edward L Barnes
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Multidisciplinary Center for Inflammatory Bowel Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
9
|
Roth D, Düll MM, Horst LJ, Lindemann A, Malzer X, Koop K, Zundler S, Vetter M, Jefremow A, Atreya R, Geppert C, Weidemann S, Waldner MJ, Dietrich P, Günther C, Munoz LE, Herrmann M, Scheffold A, Neurath MF, Siebler J, Schramm C, Kremer AE, Leppkes M. Integrin αVβ6: Autoantigen and Driver of Epithelial Remodeling in Colon and Bile Ducts in Primary Sclerosing Cholangitis and Inflammatory Bowel Disease. J Crohns Colitis 2025; 19:jjae131. [PMID: 39212221 DOI: 10.1093/ecco-jcc/jjae131] [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: 03/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Recently, autoantibodies directed against the epithelial adhesion protein integrin αVβ6 have been identified that are strongly associated with ulcerative colitis (UC). We aimed to elucidate whether anti-integrin αVβ6 (anti-αVβ6) is present in primary sclerosing cholangitis (PSC), its associated inflammatory bowel disease, or other cholestatic liver diseases and their persistence after proctocolectomy. METHODS We detected anti-αVβ6 by an enzyme-linked immunosorbent assay in sera collected at 2 German tertiary centers, including healthy controls (N = 62), UC (N = 36), Crohn's disease (CD, N = 65), PSC-inflammatory bowel diseases (IBD) (78 samples from N = 41 patients), PSC without IBD (PSC, 41 samples from N = 18 patients), primary biliary cholangitis (PBC, N = 24), autoimmune hepatitis (AIH, N = 32), secondary sclerosing cholangitis (SSC, N = 12), and metabolic dysfunction-associated steatotic liver disease (MASLD, N = 24). In addition, sera after proctocolectomy were studied (44 samples/N = 10 patients). Immunofluorescent analyses were performed in tissue samples from liver, large bile duct from surgical resections, and colon of PSC patients. RESULTS Anti-αVβ6 occurred in 91% of UC, 17% of CD, 73% of PSC-IBD, 39% of PSC, 4% of PBC, 14% of AIH, and 0% of healthy controls, SSC, or MASLD. Integrin αVβ6 is selectively expressed in disease-associated epithelia of both bile duct and colon. Anti-αVβ6 levels correlate moderately with intestinal disease activity in PSC-IBD, but only weakly with biliary disease. CONCLUSIONS Anti-αVβ6 frequently occurs in patients suffering from PSC, especially in PSC-IBD. Anti-αVβ6 levels positively correlate to IBD activity in PSC-IBD, but may also occur in the absence of clinically manifest IBD in PSC.
Collapse
Affiliation(s)
- Dominik Roth
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Miriam M Düll
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ludwig J Horst
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Aylin Lindemann
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xenia Malzer
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kristina Koop
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marcel Vetter
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - André Jefremow
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sören Weidemann
- Institute of Pathology, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Peter Dietrich
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander-University Erlangen-Nürnberg, Germany
| | - Claudia Günther
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis E Munoz
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Markus F Neurath
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jürgen Siebler
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christoph Schramm
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas E Kremer
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Hepatology and Gastroenterology, University Hospital Zürich and University Zürich, Zürich, Switzerland
| | - Moritz Leppkes
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
10
|
Fischer S, Neurath MF. [Update on the pathophysiology, prediction and prevention of inflammatory bowel diseases]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:137-145. [PMID: 39833377 DOI: 10.1007/s00108-024-01838-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND The pathophysiology of inflammatory bowel diseases is not fully understood. In a staged model by the European Crohn's and Colitis Organization (ECCO) regarding disease development, it is assumed that there is a population at risk for manifestation of disease following subtle changes over time. OBJECTIVE This work aims to summarize the current state of knowledge regarding the pathophysiology, prediction and prevention of chronic inflammatory bowel diseases. MATERIALS AND METHODS Selective literature research via PubMed. RESULTS Several genetic, biochemical, and microbiome scores have the potential to identify individuals at increased risk of developing inflammatory bowel disease, possibly up to a decade before onset. DISCUSSION The growing knowledge regarding the pathogenesis of inflammatory bowel diseases makes prediction before disease onset a possible future diagnostic goal. Hypothetically, early changes before the disease manifests could be reversible and may be amenable to prevention programs.
Collapse
Affiliation(s)
- Sarah Fischer
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Deutschland.
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Deutschland.
| | - Markus F Neurath
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Deutschland
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Deutschland
| |
Collapse
|
11
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [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: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
12
|
Okabe M, Yamamoto S, Shiokawa M, Hisamatsu T, Yamazaki H, Nakanishi R, Hamada K, Kitamoto H, Kuwada T, Uza N, Sakatani A, Fujii T, Ohno M, Matsuura M, Shibuya T, Ohmiya N, Ooi M, Hoshi N, Moriya K, Tsuchiya K, Yamaguchi Y, Kunisaki R, Takahara M, Takagi T, Takehara T, Hirai F, Kakimoto K, Esaki M, Nakase H, Kinjo F, Torisu T, Kanmura S, Narimatsu K, Matsuoka K, Hiraga H, Yokoyama K, Honzawa Y, Naganuma M, Saruta M, Kodama Y, Chiba T, Seno H. Anti-integrin αvβ6 antibody as a biomarker for diagnosing ulcerative colitis: a nationwide multicenter validation study. J Gastroenterol 2025; 60:86-95. [PMID: 39607498 PMCID: PMC11717824 DOI: 10.1007/s00535-024-02176-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: 07/03/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND A serum biomarker for diagnosing ulcerative colitis (UC) remains to be established. Although we recently reported an anti-integrin αvβ6 antibody (V6 Ab) for diagnosing UC with high sensitivity and specificity, no large-scale validation study exists. This study aimed to validate the diagnostic value of V6 Ab for UC using a nationwide multicenter cohort study. METHODS We measured V6 Ab titers in patients definitively diagnosed with UC, Crohn's disease (CD), or other gastrointestinal disorders (OGDs). The primary outcome was the diagnostic value of V6 Ab. Secondary outcomes were factors associated with false-negative results in patients with UC and false-positive results in patients without UC and the heterogeneity of the diagnostic value of V6 Ab among the participating facilities. RESULTS We enrolled 1241, 796, and 206 patients with UC, CD, and OGD, respectively, from 28 Japanese high-volume referral centers. The diagnostic sensitivity of V6 Ab for UC was 87.7%, and its specificities for CD and OGDs were 82.0% and 87.4%, respectively. Multivariable logistic regression analysis showed that false-negative results were associated with older age at the time of sample collection, current smokers, lower partial Mayo score, and not receiving advanced therapies in patients with UC, and false-positive results were associated with colonic CD in patients with CD. No factor was associated with false-positive results in patients with OGDs. There were no significant differences in the diagnostic value of V6 Ab among the centers. CONCLUSIONS The diagnostic value of V6 Ab for UC was validated in the large-scale nationwide multicenter study.
Collapse
Affiliation(s)
- Makoto Okabe
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shuji Yamamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Tadakazu Hisamatsu
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Mitaka-shi, Tokyo, 181-8611, Japan.
| | - Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Risa Nakanishi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kensuke Hamada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroki Kitamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Aki Sakatani
- Division of Gastroenterology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, 078-8510, Japan
| | - Toshimitsu Fujii
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Masashi Ohno
- Department of Medicine, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Minoru Matsuura
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Tomoyoshi Shibuya
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Naoki Ohmiya
- Department of Advanced Endoscopy, Fujita Health University School of Medicine, Toyoake, 470-1192, Japan
| | - Makoto Ooi
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Kei Moriya
- Department of Gastroenterology, Nara Prefecture General Medical Center, Nara, 630-8581, Japan
| | - Kiichiro Tsuchiya
- Department of Gastroenterology, Institute of Medicine, University of Tsukuba, Tsukuba, 305-8576, Japan
| | - Yoshiharu Yamaguchi
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, 480-1195, Japan
| | - Reiko Kunisaki
- Inflammatory Bowel Disease Center, Yokohama City University Medical Center, Yokohama, 232-0024, Japan
| | - Masahiro Takahara
- Department of Gastroenterology and Hepatology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Fumihito Hirai
- Department of Gastroenterology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kazuki Kakimoto
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, 569-8686, Japan
| | - Motohiro Esaki
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Fukunori Kinjo
- Center for Gastroenterology, Urasoe General Hospital, Urasoe, 901-2102, Japan
| | - Takehiro Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shuji Kanmura
- Division of Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8520, Japan
| | - Kazuyuki Narimatsu
- Department of Internal Medicine, National Defence Medical College, Tokorozawa, 359-8513, Japan
| | - Katsuyoshi Matsuoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Toho University Sakura Medical Center, Sakura, 285-8741, Japan
| | - Hiroto Hiraga
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8563, Japan
| | - Kaoru Yokoyama
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, 252-0375, Japan
| | - Yusuke Honzawa
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Osaka, 573-1010, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Osaka, 573-1010, Japan
| | - Masayuki Saruta
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kansai Electric Power Hospital, Fukushima-ku, Osaka, 553-0003, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
13
|
Ghasempour S, Warner N, Guan R, Rodari MM, Ivanochko D, Whittaker Hawkins R, Marwaha A, Nowak JK, Liang Y, Mulder DJ, Stallard L, Li M, Yu DD, Pluthero FG, Batura V, Zhao M, Siddiqui I, Upton JE, Hulst JM, Kahr WH, Mendoza-Londono R, Charbit-Henrion F, Hoefsloot LH, Khiat A, Moreira D, Trindade E, Espinheira MDC, Pinto Pais I, Weerts MJ, Douben H, Kotlarz D, Snapper SB, Klein C, Dowling JJ, Julien JP, Joosten M, Cerf-Bensussan N, Freeman SA, Parlato M, van Ham TJ, Muise AM. Human ITGAV variants are associated with immune dysregulation, brain abnormalities, and colitis. J Exp Med 2024; 221:e20240546. [PMID: 39526957 PMCID: PMC11554753 DOI: 10.1084/jem.20240546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/16/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Integrin heterodimers containing an Integrin alpha V subunit are essential for development and play critical roles in cell adhesion and signaling. We identified biallelic variants in the gene coding for Integrin alpha V (ITGAV) in three independent families (two patients and four fetuses) that either caused abnormal mRNA and the loss of functional protein or caused mistargeting of the integrin. This led to eye and brain abnormalities, inflammatory bowel disease, immune dysregulation, and other developmental issues. Mechanistically, the reduction of functional Integrin αV resulted in the dysregulation of several pathways including TGF-β-dependent signaling and αVβ3-regulated immune signaling. These effects were confirmed using immunostaining, RNA sequencing, and functional studies in patient-derived cells. The genetic deletion of itgav in zebrafish recapitulated patient phenotypes including retinal and brain defects and the loss of microglia in early development as well as colitis in juvenile zebrafish with reduced SMAD3 expression and transcriptional regulation. Taken together, the ITGAV variants identified in this report caused a previously unknown human disease characterized by brain and developmental defects in the case of complete loss-of-function and atopy, neurodevelopmental defects, and colitis in cases of incomplete loss-of-function.
Collapse
Affiliation(s)
- Sina Ghasempour
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Neil Warner
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Rei Guan
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Marco M. Rodari
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Canada
| | | | - Ashish Marwaha
- Division of Genetics, Department of Medical Genetics, University of Calgary, Alberta Children’s Hospital, Calgary, Canada
| | - Jan K. Nowak
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Yijing Liang
- Center for Computational Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Daniel J. Mulder
- Department of Pediatrics, Gastrointestinal Diseases Research Unit, Queen’s University, Kingston, Canada
| | - Lorraine Stallard
- National Centre for Pediatric Gastroenterology, Children’s Health Ireland, Dublin, Ireland
| | - Michael Li
- Center for Computational Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Daniel D. Yu
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Fred G. Pluthero
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Vritika Batura
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Mo Zhao
- Genetics and Genome Biology, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Iram Siddiqui
- Division of Pathology, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Julia E.M. Upton
- Division of Immunology and Allergy, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Jessie M. Hulst
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, Canada
| | - Walter H.A. Kahr
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Roberto Mendoza-Londono
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Fabienne Charbit-Henrion
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
- Genomic Medicine for Rare Diseases, Necker-Enfants Malades Hospital, Paris, France
| | - Lies H. Hoefsloot
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Anis Khiat
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Diana Moreira
- Consulta de Imunodeficiências Primárias, Serviço de Pediatria, Centro Hospitalar Vila Nova de Gaia e Espinho, Vila Nova de Gaia, Portugal
| | - Eunice Trindade
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Maria do Céu Espinheira
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Isabel Pinto Pais
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Marjolein J.A. Weerts
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hannie Douben
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Daniel Kotlarz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Child and Adolescent Health, Munich Site, Munich, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston, Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Child and Adolescent Health, Munich Site, Munich, Germany
| | - James J. Dowling
- Genetics and Genome Biology, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Marieke Joosten
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Spencer A. Freeman
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Marianna Parlato
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Aleixo M. Muise
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
14
|
Bronze S, Agrawal M, Colombel JF, Torres J, Ungaro RC. Review article: Prevention of inflammatory bowel disease-The path forward. Aliment Pharmacol Ther 2024; 60:1166-1175. [PMID: 39403049 DOI: 10.1111/apt.18263] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The possibility of preventing inflammatory bowel disease (IBD) is becoming more plausible due to advances in understanding preclinical disease and successful prevention trials in other immune-mediated diseases, such as type 1 diabetes and rheumatoid arthritis. However, before that possibility becomes reality, several efforts need to occur in parallel and in a coordinated way. AIM To propose some critical steps necessary for advancing the field of IBD prediction and prevention. METHODS We reviewed the current literature to identify the necessary steps toward a preventive strategy for IBD. RESULTS The first step should determine the most robust predictive biomarkers and validate them across independent cohorts, creating a multidimensional predictive tool. The second step is to gain a better understanding of the preferences of first-degree relatives and people at risk for IBD, informing the implementation of screening and preventive strategies. Third, these efforts should contribute to the development of high-risk clinics and establish the necessary networks for disease prevention trials. CONCLUSIONS Advancing the field of IBD prediction and prevention will require a multifaceted approach, integrating biomarker discovery, understanding patient preferences, and establishing infrastructure for a collaborative network to support the practical implementation of IBD prevention strategies.
Collapse
Affiliation(s)
- Sérgio Bronze
- Gastroenterology and Hepatology Department, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal
- Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisbon, Portugal
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Manasi Agrawal
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jean-Frédéric Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joana Torres
- Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisbon, Portugal
- Division of Gastroenterology, Hospital da luz Lisboa, Lisbon, Portugal
- Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Ryan C Ungaro
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
15
|
Swaminathan A, Day AS, Sparrow MP, Peyrin-Biroulet L, Siegel CA, Gearry RB. Review article: Measuring disease severity in inflammatory bowel disease - Beyond treat to target. Aliment Pharmacol Ther 2024; 60:1176-1199. [PMID: 39403053 DOI: 10.1111/apt.18231] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) follows a heterogenous disease course and predicting a patient's prognosis is challenging. There is a wide burden of illness in IBD and existing tools measure disease activity at a snapshot in time. Comprehensive assessment of IBD severity should incorporate disease activity, prognosis, and the impacts of disease on a patient. This review investigates the concept of disease severity in adults with IBD to highlight key components contributing to this. METHODS To perform this narrative review, a Medline search was conducted for full-text articles available at 1st March 2024 using search terms which encompassed disease activity assessment, disease severity, prognosis, natural history of Crohn's disease (CD) and ulcerative colitis (UC), and the burden of IBD. RESULTS Current methods of disease assessment in IBD have evolved from a focus on the burden of symptoms to one that includes inflammatory targets, genetic, serological, and proteomic profiles, and assessments of quality-of-life (QoL), disability, and psychosocial health. Longitudinal studies of IBD suggest that the burden of illness is driven by disease phenotype, clinical markers of complicated disease course (previous intestinal resection, corticosteroid use, perianal disease in CD, recent hospitalisations in UC), gut inflammation, and the impact of IBD on the patient. CONCLUSIONS Disease severity in IBD can be difficult to conceptualise due to the multitude of factors that contribute to IBD outcomes. Measurement of IBD severity may better encapsulate the full burden of illness rather than gut inflammation alone at a single timepoint and may be associated with longitudinal outcomes.
Collapse
Affiliation(s)
- Akhilesh Swaminathan
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| | - Andrew S Day
- Department of Paediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - Miles P Sparrow
- Department of Gastroenterology, Alfred Health and School of Translational Medicine, Monash University, Australia
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, Vandoevre-les-Nancy, France
- Department of Gastroenterology, INFINY Institute, FHU-CURE, INSERM NGERE, Nancy University Hospital, Vandoeuvre-les-Nancy, France
- Groupe Hospitalier privé Ambroise Paré - Hartmann, Paris IBD Center, Neuilly sur Seine, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Corey A Siegel
- Center for Digestive Health, Section of Gastroenterology and Hepatology, Dartmouth Hitchcock Medical Centre, Lebanon, New Hampshire, USA
| | - Richard B Gearry
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| |
Collapse
|
16
|
Yamamoto S, Kuwada T, Shiokawa M, Kitamoto H, Okabe M, Seno H. Anti-Integrin αvβ6 Antibody Titer as a Predictive Biomarker of Future Treatment Escalation in Patients With Ulcerative Colitis. GASTRO HEP ADVANCES 2024; 4:100582. [PMID: 39926205 PMCID: PMC11803832 DOI: 10.1016/j.gastha.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 02/11/2025]
Affiliation(s)
- Shuji Yamamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroki Kitamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Okabe
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Bloemen H, Livanos AE, Martins A, Dean R, Bravo AC, Bourgonje AR, Tankelevich M, Herb J, Cho J, Santos AA, Rodrigues CMP, Petralia F, Colombel JF, Bowlus CL, Schiano T, Torres J, Levy C, Mehandru S. Anti-integrin αvβ6 Autoantibodies are Increased in Primary Sclerosing Cholangitis Patients With Concomitant Inflammatory Bowel Disease and Correlate With Liver Disease Severity. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00969-8. [PMID: 39490950 PMCID: PMC12022142 DOI: 10.1016/j.cgh.2024.10.005] [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: 05/11/2024] [Revised: 09/13/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND & AIMS Anti-integrin αvβ6 autoantibodies (anti-αvβ6) are found in more than 50% of individuals with ulcerative colitis (UC). We aimed to determine the prevalence of anti-αvβ6 in patients with primary sclerosing cholangitis (PSC) and their association with liver disease severity. METHODS Four cohorts of pre-liver transplant patients with PSC were recruited. Patients with inflammatory bowel disease (IBD) and healthy controls (HCs) served as comparators. Total IgG and anti-αvβ6 levels were measured using enzyme-linked immunosorbent assay. Olink inflammation panel was run on a subset of samples. Multivariable linear regression analysis was performed to assess the association between anti-αvβ6 and indices of liver disease severity. RESULTS A total of 137 patients with PSC (including 76 with PSC-UC, 33 with PSC-Crohn's disease (CD), and 28 with PSC alone) and 160 controls (including 91 with IBD and 69 HCs) were enrolled. Anti-αvβ6 levels were significantly higher in PSC-UC and PSC-CD compared with PSC alone (P < .0001 and P < .003) and HCs (P < .0001 and P < .0001). However, anti-αvβ6 levels in PSC alone were not increased compared with HCs. In patients with PSC-IBD, anti-αvβ6 levels correlated with markers of liver disease severity, including alkaline phosphatase level (r = 0.32; P = .004), the revised Mayo PSC risk score (r = 0.25; P = .02), and liver stiffness measurement (r = 0.43; P = .008) after adjusting for age, gender, race/ethnicity, and IBD subtype. Additionally, anti-αvβ6 levels were associated with markers of systemic inflammation and tissue remodeling. CONCLUSION Anti-αvβ6 autoantibodies identify a subset of patients with PSC with concomitant IBD.
Collapse
Affiliation(s)
- Hannah Bloemen
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexandra E Livanos
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Adrielly Martins
- Schiff Center for Liver Diseases, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Richard Dean
- Division of Gastroenterology and Hepatology, University of California Davis School of Medicine, Sacramento, California
| | | | - Arno R Bourgonje
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Tankelevich
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jake Herb
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judy Cho
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - André Anastácio Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher L Bowlus
- Division of Gastroenterology and Hepatology, University of California Davis School of Medicine, Sacramento, California
| | - Thomas Schiano
- Recanati/Miller Transplantation Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joana Torres
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal; Faculty of Medicine, Universidade de Lisboa, Lisbon, Portugal; Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
| | - Cynthia Levy
- Schiff Center for Liver Diseases, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Division of Digestive Health and Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida.
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
18
|
Wangchuk P, Yeshi K, Loukas A. Ulcerative colitis: clinical biomarkers, therapeutic targets, and emerging treatments. Trends Pharmacol Sci 2024; 45:892-903. [PMID: 39261229 DOI: 10.1016/j.tips.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024]
Abstract
Ulcerative colitis (UC) is one of the two forms of inflammatory bowel disease. It affects 5 million people globally, and is a chronic and recurring inflammation of the gastrointestinal tract with clinical presentation of abdominal pain, chronic diarrhea, rectal bleeding, and weight loss. The cause and the etiology of UC remain poorly understood. There is no cure and no 'gold standard diagnostic' for UC. The existing treatments are ineffective, and UC patients have a lower life expectancy with a risk of colorectal cancer. Recent studies in pathophysiology, clinical presentation, and biomarkers have significantly improved our understanding of UC. In this review we summarize recent advances in identifying novel clinical biomarkers, diagnostics, treatment targets, and emerging therapeutics. These insights are expected to assist in developing effective treatments for UC.
Collapse
Affiliation(s)
- Phurpa Wangchuk
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns campus, James Cook University, QLD 4878, Australia; Australian Institute of Tropical Health and Medicine (AITHM), Cairns campus, James Cook University, QLD 4878, Australia.
| | - Karma Yeshi
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns campus, James Cook University, QLD 4878, Australia; Australian Institute of Tropical Health and Medicine (AITHM), Cairns campus, James Cook University, QLD 4878, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine (AITHM), Cairns campus, James Cook University, QLD 4878, Australia
| |
Collapse
|
19
|
Griffin H, Ceron-Gutierrez L, Gharahdaghi N, Ebrahimi S, Davies S, Loo PS, Szabo A, Williams E, Mukhopadhyay A, McLoughlin L, Irwin S, Travis S, Klenerman P, Bunn S, Cant AJ, Hambleton S, Uhlig HH, Doffinger R. Neutralizing Autoantibodies against Interleukin-10 in Inflammatory Bowel Disease. N Engl J Med 2024; 391:434-441. [PMID: 39083772 PMCID: PMC7616361 DOI: 10.1056/nejmoa2312302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
We discovered high-titer neutralizing autoantibodies against interleukin-10 in a child with infantile-onset inflammatory bowel disease (IBD), a phenocopy of inborn errors of interleukin-10 signaling. After B-cell-depletion therapy and an associated decrease in the anti-interleukin-10 titer, conventional IBD therapy could be withdrawn. A second child with neutralizing anti-interleukin-10 autoantibodies had a milder course of IBD and has been treated without B-cell depletion. We conclude that neutralizing anti-interleukin-10 autoantibodies may be a causative or modifying factor in IBD, with potential implications for therapy. (Funded by the National Institute for Health and Care Research and others.).
Collapse
Affiliation(s)
- Helen Griffin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Lourdes Ceron-Gutierrez
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Nima Gharahdaghi
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Soraya Ebrahimi
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Sophie Davies
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Peh Sun Loo
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Andras Szabo
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Eleri Williams
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Anirban Mukhopadhyay
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Louise McLoughlin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Steven Irwin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Simon Travis
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Paul Klenerman
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Su Bunn
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Andrew J Cant
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Sophie Hambleton
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Holm H Uhlig
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Rainer Doffinger
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| |
Collapse
|
20
|
Lopes EW, Turpin W, Croitoru K, Colombel JF, Torres J. Prediction and Prevention of Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00597-4. [PMID: 38996831 DOI: 10.1016/j.cgh.2024.05.047] [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: 03/11/2024] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 07/14/2024]
Affiliation(s)
- Emily W Lopes
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Frederic Colombel
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joana Torres
- Division of Gastrenterology, Hospital da Luz, Lisboa, Portugal; Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal; Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
21
|
Mestrovic A, Perkovic N, Bozic D, Kumric M, Vilovic M, Bozic J. Precision Medicine in Inflammatory Bowel Disease: A Spotlight on Emerging Molecular Biomarkers. Biomedicines 2024; 12:1520. [PMID: 39062093 PMCID: PMC11274502 DOI: 10.3390/biomedicines12071520] [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: 05/31/2024] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammatory bowel diseases (IBD) remain challenging in terms of understanding their causes and in terms of diagnosing, treating, and monitoring patients. Modern diagnosis combines biomarkers, imaging, and endoscopic methods. Common biomarkers like CRP and fecal calprotectin, while invaluable tools, have limitations and are not entirely specific to IBD. The limitations of existing markers and the invasiveness of endoscopic procedures highlight the need to discover and implement new markers. With an ideal biomarker, we could predict the risk of disease development, as well as the possibility of response to a particular therapy, which would be significant in elucidating the pathogenesis of the disease. Recent research in the fields of machine learning, proteomics, epigenetics, and gut microbiota provides further insight into the pathogenesis of the disease and is also revealing new biomarkers. New markers, such as BAFF, PGE-MUM, oncostatin M, microRNA panels, αvβ6 antibody, and S100A12 from stool, are increasingly being identified, with αvβ6 antibody and oncostatin M being potentially close to being presented into clinical practice. However, the specificity of certain markers still remains problematic. Furthermore, the use of expensive and less accessible technology for detecting new markers, such as microRNAs, represents a limitation for widespread use in clinical practice. Nevertheless, the need for non-invasive, comprehensive markers is becoming increasingly important regarding the complexity of treatment and overall management of IBD.
Collapse
Affiliation(s)
- Antonio Mestrovic
- Department of Gastroenterology, University Hospital of Split, Spinciceva 2, 21000 Split, Croatia; (A.M.); (N.P.); (D.B.)
| | - Nikola Perkovic
- Department of Gastroenterology, University Hospital of Split, Spinciceva 2, 21000 Split, Croatia; (A.M.); (N.P.); (D.B.)
| | - Dorotea Bozic
- Department of Gastroenterology, University Hospital of Split, Spinciceva 2, 21000 Split, Croatia; (A.M.); (N.P.); (D.B.)
| | - Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia;
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| | - Marino Vilovic
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia;
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia;
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| |
Collapse
|
22
|
Ashton JJ, Satsangi J, Uhlig HH. Intestinal Inflammation and Extraintestinal Disease: Understanding Dynamic Risk. Gastroenterology 2024; 167:205-208. [PMID: 38604541 DOI: 10.1053/j.gastro.2024.03.040] [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: 03/06/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Affiliation(s)
- James J Ashton
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, United Kingdom; Department of Paediatric Gastroenterology, Southampton Children's Hospital, Southampton, United Kingdom
| | - Jack Satsangi
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom; Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Department of Paediatrics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
23
|
Hong SM, Baek DH. Diagnostic Procedures for Inflammatory Bowel Disease: Laboratory, Endoscopy, Pathology, Imaging, and Beyond. Diagnostics (Basel) 2024; 14:1384. [PMID: 39001273 PMCID: PMC11241288 DOI: 10.3390/diagnostics14131384] [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: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Diagnosing inflammatory bowel disease (IBD) can often be challenging, and differentiating between Crohn's disease and ulcerative colitis can be particularly difficult. Diagnostic procedures for IBD include laboratory tests, endoscopy, pathological tests, and imaging tests. Serological and stool tests can be easily performed in an outpatient setting and provide critical diagnostic clues. Although endoscopy is an invasive procedure, it offers essential diagnostic information and allows for tissue biopsy and therapeutic procedures. Video capsule endoscopy and device-assisted enteroscopy are endoscopic procedures used to evaluate the small bowel. In addition to endoscopy, magnetic resonance imaging, computed tomography, and ultrasound (US) are valuable tools for small bowel assessment. Among these, US is noninvasive and easily utilized, making its use highly practical in daily clinical practice. Endoscopic biopsy aids in the diagnosis of IBD and is crucial for assessing the histological activity of the disease, facilitating a thorough evaluation of disease remission, and aiding in the development of treatment strategies. Recent advances in artificial intelligence hold promise for enhancing various aspects of IBD management, including diagnosis, monitoring, and precision medicine. This review compiles current procedures and promising future tools for the diagnosis of IBD, providing comprehensive insights.
Collapse
Affiliation(s)
- Seung Min Hong
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Dong Hoon Baek
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| |
Collapse
|
24
|
Rodríguez-Lago I, Aduna M, Ramírez de la Piscina P, Merino O, Carrascosa J, Higuera R, Maíz A, Zapata E, Cabriada JL, Barreiro-de Acosta M. Transmural cross-sectional findings and bowel damage assessment in preclinical Crohn's disease: a case-control study. Int J Colorectal Dis 2024; 39:92. [PMID: 38871954 PMCID: PMC11176094 DOI: 10.1007/s00384-024-04660-5] [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] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Crohn's disease (CD) is a progressive disorder leading to cumulative bowel damage. The Lémann index is a validated tool that can help in monitoring the progression of the disease and evaluating the effectiveness of different therapies. Our aim was to describe the main radiological findings in incidentally diagnosed CD and to evaluate bowel damage in this subgroup compared to patients diagnosed at later stages. METHODS Patients with an incidental diagnosis of CD during the colorectal cancer screening program were compared to controls with a CD cohort diagnosed after symptomatic onset and matched 1:1 by disease extent. All cross-sectional examinations were centrally read, performing a descriptive analysis of the main findings and calculation of Lémann index. RESULTS Thirty-eight patients were included: 19 with preclinical CD (median age 55 years (IQR, 54-62), 53% male, 74% non-smokers; 74% B1 and 26% B2) and 19 matched-controls with symptomatic CD. In those with preclinical CD, the most frequent transmural findings on MRE were contrast enhancement (79%), wall thickening (79%), followed by lymphadenopathy (68%), edema (42%), and increased vascularity (42%). Among those with strictures, controls showed a higher rate of preestenotic dilation (100% vs. 0%, p = 0.01). Bowel damage assessment revealed no statistically significant differences in the Lémann index between preclinical CD and controls (p = 0.95). A statistically significant higher score in the colonic/rectum score was observed (p = 0.014). CONCLUSION Patients with preclinical CD demonstrate similar radiological findings and degree of bowel damage as new-onset symptomatic CD.
Collapse
Affiliation(s)
- Iago Rodríguez-Lago
- IBD Unit, Gastroenterology Department, Hospital Universitario de Galdakao-Usansolo, Galdakao, Spain.
- Biobizkaia Health Research Institute, Galdakao, Spain.
- School of Medicine, Universidad de Deusto, Bilbao, Spain.
| | - Marta Aduna
- School of Medicine, Universidad de Deusto, Bilbao, Spain
- OSATEK, Galdakao, Spain
| | | | - Olga Merino
- Gastroenterology Department, Hospital Universitario de Cruces, Barakaldo, Spain
| | - Juan Carrascosa
- Gastroenterology Department, Hospital de Zumárraga, Zumárraga, Spain
| | - Rebeca Higuera
- Gastroenterology Department, Hospital San Eloy, Barakaldo, Spain
| | - Ainara Maíz
- Gastroenterology Department, Hospital Universitario Donostia, Donostia, Spain
| | - Eva Zapata
- Gastroenterology Department, Hospital de Mendaro, Mendaro, Spain
| | - José Luis Cabriada
- IBD Unit, Gastroenterology Department, Hospital Universitario de Galdakao-Usansolo, Galdakao, Spain
| | - Manuel Barreiro-de Acosta
- Gastroenterology Department, Hospital Clínico Universitario de Santiago, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
25
|
Prasad S, Cross RK, Monroe MB, Dolinger MT, Motte R, Hong S, Stidham RW, Kumar N, Levine D, Larijani A, Simone A, Chachu KA, Wyborski R, Heller CA, Moss AC, Schwerbrock NMJ, Selaru FM. Challenges in IBD Research 2024: Novel Technologies. Inflamm Bowel Dis 2024; 30:S30-S38. [PMID: 38778625 DOI: 10.1093/ibd/izae082] [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: 03/13/2024] [Indexed: 05/25/2024]
Abstract
Novel technology is one of the five focus areas of the Challenges in Inflammatory Bowel Disease (IBD) Research 2024 document. Building off the Challenges in IBD Research 2019 document, the Foundation aims to provide a comprehensive overview of current gaps in IBD research and deliver actionable approaches to address them with a focus on how these gaps can lead to advancements in interception, remission, and restoration for these diseases. The document is the result of a multidisciplinary collaboration from scientists, clinicians, patients, and funders and represents a valuable resource for patient-centric research prioritization. Specifically, the Novel Technologies section focuses on addressing key research gaps to enable interception and improve remission rates in IBD. This includes testing predictions of disease onset and progression, developing novel technologies tailored to specific phenotypes, and facilitating collaborative translation of science into diagnostics, devices, and therapeutics. Proposed priority actions outlined in the document include real-time measurement of biological changes preceding disease onset, more effective quantification of fibrosis, exploration of technologies for local treatment of fistulas, and the development of drug delivery platforms for precise, location-restricted therapies. Additionally, there is a strong emphasis on fostering collaboration between various stakeholders to accelerate progress in IBD research and treatment. Addressing these research gaps necessitates the exploration and implementation of bio-engineered novel technologies spanning a spectrum from materials to systems. By harnessing innovative ideas and technologies, there's a collective effort to enhance patient care and outcomes for individuals affected by IBD.
Collapse
Affiliation(s)
- Shalini Prasad
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Raymond K Cross
- Director of the Inflammatory Bowel Disease Program, University of Maryland School of Medicine, Maryland, MD, USA
| | - Mary Beth Monroe
- Department of Biomedical and Chemical Engineering BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Michael T Dolinger
- Icahn School of Medicine at Mount Sinai, Division of Pediatric Gastroenterology, New York, NY, USA
| | - Rachel Motte
- TISSIUM, 74 Rue du Faubourg Saint-Antoine, Paris, France
| | - Sungmo Hong
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Ryan W Stidham
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Narendra Kumar
- Department of Pharmaceutical Science, ILR-College of Pharmacy, Texas A&M University, TX, USA
| | - Deborah Levine
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Anthony Larijani
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Ashley Simone
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Karen A Chachu
- Department of Medicine, Division of Gastroenterology, Duke University School of Medicine, Durham, NC, USA
| | | | - Caren A Heller
- Members of the Crohn's & Colitis Foundation, New York, NY, USA
| | - Alan C Moss
- Members of the Crohn's & Colitis Foundation, New York, NY, USA
| | | | - Florin M Selaru
- Division of Gastroenterology, Oncology and Biomedical Engineering, Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
26
|
Syed S, Boland BS, Bourke LT, Chen LA, Churchill L, Dobes A, Greene A, Heller C, Jayson C, Kostiuk B, Moss A, Najdawi F, Plung L, Rioux JD, Rosen MJ, Torres J, Zulqarnain F, Satsangi J. Challenges in IBD Research 2024: Precision Medicine. Inflamm Bowel Dis 2024; 30:S39-S54. [PMID: 38778628 DOI: 10.1093/ibd/izae084] [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: 03/15/2024] [Indexed: 05/25/2024]
Abstract
Precision medicine is part of 5 focus areas of the Challenges in IBD Research 2024 research document, which also includes preclinical human IBD mechanisms, environmental triggers, novel technologies, and pragmatic clinical research. Building on Challenges in IBD Research 2019, the current Challenges aims to provide a comprehensive overview of current gaps in inflammatory bowel diseases (IBDs) research and deliver actionable approaches to address them with a focus on how these gaps can lead to advancements in interception, remission, and restoration for these diseases. The document is the result of multidisciplinary input from scientists, clinicians, patients, and funders, and represents a valuable resource for patient-centric research prioritization. In particular, the precision medicine section is focused on the main research gaps in elucidating how to bring the best care to the individual patient in IBD. Research gaps were identified in biomarker discovery and validation for predicting disease progression and choosing the most appropriate treatment for each patient. Other gaps were identified in making the best use of existing patient biosamples and clinical data, developing new technologies to analyze large datasets, and overcoming regulatory and payer hurdles to enable clinical use of biomarkers. To address these gaps, the Workgroup suggests focusing on thoroughly validating existing candidate biomarkers, using best-in-class data generation and analysis tools, and establishing cross-disciplinary teams to tackle regulatory hurdles as early as possible. Altogether, the precision medicine group recognizes the importance of bringing basic scientific biomarker discovery and translating it into the clinic to help improve the lives of IBD patients.
Collapse
Affiliation(s)
- Sana Syed
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
- Patient representative for Crohn's & Colitis Foundation, New York, NY, USA
| | - Brigid S Boland
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Lauren T Bourke
- Precision Medicine Drug Development, Early Respiratory and Immunology, AstraZeneca, Boston, MA, USA
| | - Lea Ann Chen
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Laurie Churchill
- Leona M. and Harry B. Helmsley Charitable Trust, New York, NY, USA
| | | | - Adam Greene
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - Alan Moss
- Crohn's & Colitis Foundation, New York, NY, USA
| | | | - Lori Plung
- Patient representative for Crohn's & Colitis Foundation, New York, NY, USA
| | - John D Rioux
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Michael J Rosen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Joana Torres
- Division of Gastroenterology, Hospital Beatriz Ângelo, Hospital da Luz, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Fatima Zulqarnain
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Jack Satsangi
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Jelinsky S, Lee I, Monetti M, Breitkopf S, Martz F, Kongala R, Culver J, Vo V, Xue L, Gieseck R, Dickinson C, Kasaian M, Lord JD. Proteomic Differences in Colonic Epithelial Cells in Ulcerative Colitis Have an Epigenetic Basis. GASTRO HEP ADVANCES 2024; 3:830-841. [PMID: 39280905 PMCID: PMC11401595 DOI: 10.1016/j.gastha.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/30/2024] [Indexed: 09/18/2024]
Abstract
Background and Aims The colonic epithelium serves as both a barrier to lumenal contents and a gatekeeper of inflammatory responses. In ulcerative colitis (UC), epithelial dysfunction is a core feature, but little is known about the cellular changes that may underlie disease pathology. We therefore evaluated how the chromatin epigenetics and proteome of epithelial cells differs between health and UC. Methods We sorted live CD326+ epithelial cells from colon biopsies of healthy control (HC) screening colonoscopy recipients and from inflamed or uninflamed colon segments of UC patients on no biologic nor immunomodulator therapy (n = 5-7 subjects per group). Cell lysates were analyzed by proteomic evaluation and nuclei were analyzed for open chromatin with assay for transposase-accessible chromatin using sequencing. Results Proteins most highly elevated in inflamed UC biopsies relative to HC were those encoded by the HLA-DRA (P = 3.1 × 10-33) and CD74 (P = 1.6 × 10-27), genes associated with antigen presentation, and the antimicrobial dual oxidase 2 (DUOX2) (P = 3.2 × 10-28) and lipocalin-2 (P = 2.2 × 10-26) genes. Conversely, the water channel aquaporin 8 was strikingly less common with inflammation (P = 1.9 × 10-18). Assay for transposase-accessible chromatin using sequencing revealed more open chromatin around the aquaporin 8 gene in HCs (P = 2.0 × 10-2) and more around the DUOX2/DUOXA2 locus in inflamed UC colon (P = 5.7 × 10-4), suggesting an epigenetic basis for differential protein expression by epithelial cells in health and disease. Conclusion Numerous differences exist between the proteome and chromatin of colonic epithelial cells in UC patients and HCs, some of which correlate to suggest specific epigenetic mechanisms regulating the epithelial proteome.
Collapse
Affiliation(s)
- Scott Jelinsky
- Department of Inflammation and Immunology, Pfizer, Cambridge, Massachusetts
| | - Isac Lee
- Department of Inflammation and Immunology, Pfizer, Cambridge, Massachusetts
| | - Mara Monetti
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts
| | | | - Flora Martz
- Translational Research Program, Benaroya Research Institute, Seattle, Washington
| | - Ramya Kongala
- Translational Research Program, Benaroya Research Institute, Seattle, Washington
| | - Jeffrey Culver
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts
| | - Vanessa Vo
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts
| | - Liang Xue
- Machine Learning and Computational Sciences, Early Clinical Development, Pfizer, Cambridge, Massachusetts
| | - Richard Gieseck
- Department of Inflammation and Immunology, Pfizer, Cambridge, Massachusetts
| | - Caitlyn Dickinson
- Department of Inflammation and Immunology, Pfizer, Cambridge, Massachusetts
| | - Marion Kasaian
- Department of Inflammation and Immunology, Pfizer, Cambridge, Massachusetts
| | - James D Lord
- Translational Research Program, Benaroya Research Institute, Seattle, Washington
| |
Collapse
|
28
|
Yang J, Huang MMC, Liang MMJW, Lei MMYC. The diagnostic performance of serum αvβ6 autoantibodies for ulcerative colitis: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 2024; 48:102317. [PMID: 38499246 DOI: 10.1016/j.clinre.2024.102317] [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: 12/25/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Currently, there is no single golden standard for diagnosing ulcerative colitis (UC). Now serum αvβ6 autoantibodies have shown promise as a diagnostic tool for UC. Here the aim was to determine the diagnostic performance of serum αvβ6 autoantibodies for UC. METHODS PubMed, the Cochrane Library, the Embase, and the Web of Science were searched comprehensively. STATA software was utilized to analyze the relevant data. RESULTS 9 studies from 6 articles with 1827 subjects were eligible. The summary sensitivity and specificity of serum αvβ6 autoantibodies to diagnose UC were 0.82 (95 % confidence interval (CI): 0.65-0.92) and 0.94 (95 % CI: 0.90-0.97) with an area under the summary receiver operating characteristic curve of 0.96 (95 % CI: 0.94-0.97). Subgroup analysis was conducted owning to substantial heterogeneity between studies (I2 = 97 % and P < 0.001). The aggregate sensitivity and specificity to diagnose UC in adults were 0.75 (95 % CI: 0.61-0.86) and 0.95 (95 % CI: 0.90-0.97), and when using a threshold of mean control+3SD, 0.80 (95 % CI: 0.60-0.91) and 0.96 (95 % CI: 0.90-0.99), respectively. Additionally, to differentiate UC from healthy participants, non-inflammatory bowel disease, and Crohn's disease, the overall specificity was 0.96, 0.88, and 0.80, respectively. CONCLUSIONS serum αvβ6 autoantibodies, as a non-invasive tool, demonstrated good diagnostic accuracy for UC. However, their application may be limited in some immune-related disorders, and further studies are needed for validation.
Collapse
Affiliation(s)
- Jiao Yang
- Department of Gastroenterology, LiuZhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - M M Cai Huang
- Department of Gastroenterology, LiuZhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - M M Jing-Wen Liang
- Department of Gastroenterology, LiuZhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - M M Yan-Chang Lei
- Department of Gastroenterology, LiuZhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China.
| |
Collapse
|
29
|
Alomari M, Chadalavada P, Afraz S, AlGhadir-AlKhalaileh M, Suarez ZK, Swartz A, Rashid M, Khazaaleh S, Cohen BL, Ur Rahman A, Alomari M. Post-hospitalization Short Versus Long Steroid Taper Strategies in Patients With Acute Severe Ulcerative Colitis: A Comparison of Clinical Outcomes. CROHN'S & COLITIS 360 2024; 6:otae025. [PMID: 38711857 PMCID: PMC11071514 DOI: 10.1093/crocol/otae025] [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] [Received: 12/25/2023] [Indexed: 05/08/2024] Open
Abstract
Background Ulcerative colitis (UC) is a chronic inflammatory colon disease characterized by relapsing flares and remission episodes. However, the optimal steroid tapering strategy in patients hospitalized for acute severe UC (ASUC) remains relatively unknown. We aim to examine the clinical outcomes in patients hospitalized for ASUC regarding variable prednisone taper regimens upon discharge. Methods We retrospectively reviewed all adult patients admitted to our facility with ASUC between 2000 and 2022. Patients were divided into 2 groups based on the duration of steroid taper on discharge (< 6 and > 6 weeks). Patients who had colectomy at index admission were excluded from the analysis. The primary outcome was rehospitalization for ASUC within 6 months of index admission. Secondary outcomes included the need for colectomy, worsening endoscopic disease extent and/or severity during the follow-up period (6 months), and a composite outcome as a surrogate of worsening disease (defined as a combination of all products above). Two-sample t-tests and Pearson's chi-square tests were used to compare the means of continuous and categorical variables, respectively. Multivariate logistic regression analysis was performed to identify independent predictors for rehospitalization with ASUC. Results A total of 215 patients (short steroid taper = 91 and long steroid taper = 124) were analyzed. A higher number of patients in the long steroid taper group had a longer disease duration since diagnosis and moderate-severe endoscopic disease activity (63.8 vs. 25.6 months, p < 0.0001, 46.8% vs. 23.1%, P = ≤ .05, respectively). Both groups had similar disease extent, prior biologic therapy, and the need for inpatient rescue therapy. At the 6-month follow-up, rates of rehospitalization with a flare of UC were comparable between the 2 groups (68.3% vs. 68.5%, P = .723). On univariate and multivariate logistic regression, escalation of steroid dose within four weeks of discharge (aOR 6.09, 95% CI: 1.82-20.3, P = .003) was noted to be the only independent predictor for rehospitalization with ASUC. Conclusions This is the first study comparing clinical outcomes between post-discharge steroid tapering regimens in hospitalized patients for ASUC. Both examined steroid taper regimens upon discharge showed comparable clinical results. Hence, we suggest a short steroid taper as a standard post-hospitalization strategy in patients following ASUC encounters. It is likely to enhance patient tolerability and reduce steroid-related adverse effects without adversely affecting outcomes.
Collapse
Affiliation(s)
- Mohammad Alomari
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Pravallika Chadalavada
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL, USA
| | - Sadaf Afraz
- Internal Medicine Department, Cleveland Clinic Florida, Weston, FL, USA
| | | | - Zoilo K Suarez
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Alec Swartz
- Internal Medicine Department, Cleveland Clinic Florida, Weston, FL, USA
| | - Mamoon Rashid
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL, USA
| | - Shrouq Khazaaleh
- Internal Medicine Department, Cleveland Clinic Fairview Hospital, Cleveland, OH, USA
| | - Benjamin L Cohen
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Asad Ur Rahman
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL, USA
| | | |
Collapse
|
30
|
Xu C, Shao J. High-throughput omics technologies in inflammatory bowel disease. Clin Chim Acta 2024; 555:117828. [PMID: 38355001 DOI: 10.1016/j.cca.2024.117828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing intestinal disease. Elucidation of the pathogenic mechanisms of IBD requires high-throughput technologies (HTTs) to effectively obtain and analyze large amounts of data. Recently, HTTs have been widely used in IBD, including genomics, transcriptomics, proteomics, microbiomics, metabolomics and single-cell sequencing. When combined with endoscopy, the application of these technologies can provide an in-depth understanding on the alterations of intestinal microbe diversity and abundance, the abnormalities of signaling pathway-mediated immune responses and functionality, and the evaluation of therapeutic effects, improving the accuracy of early diagnosis and treatment of IBD. This review comprehensively summarizes the development and advancement of HTTs, and also highlights the challenges and future directions of these technologies in IBD research. Although HTTs have made striking breakthrough in IBD, more standardized methods and large-scale dataset processing are still needed to achieve the goal of personalized medicine.
Collapse
Affiliation(s)
- Chen Xu
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Jing Shao
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China.
| |
Collapse
|
31
|
Schaub JR, Chen JY, Turner SM. Integrins in biliary injury and fibrosis. Curr Opin Gastroenterol 2024; 40:85-91. [PMID: 38190346 DOI: 10.1097/mog.0000000000000995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
PURPOSE OF REVIEW Current treatment options for cholangiopathies are severely limited and there is thus a critical need to identify and develop therapies. This review discusses the role of integrins in biliary injury and fibrosis and their potential as therapeutic targets. RECENT FINDINGS There are a diverse set of roles that integrins play in biliary injury and fibrosis. Some integrins activate TGF-β signaling or are involved in sensing of the extracellular matrix, making them attractive targets for biliary fibrosis. In recent work, autoantibodies to α v β 6 were identified in patients with PSC, supporting the relevance of this integrin in the disease. In addition, a role for α 2 β 1 in cyst formation was identified in a mouse model of polycystic liver disease. Leukocyte integrins (e.g. α E β 7 and α 4 β 7 ) contribute to lymphocyte trafficking, making them potential targets for biliary inflammation; however, this has not yet translated to the clinic. SUMMARY While all members of the same family of proteins, integrins have diverse roles in the pathogenesis of biliary disease. Targeting one or multiple of these integrins may slow or halt the progression of biliary injury and fibrosis by simultaneously impacting different pathologic cells and processes.
Collapse
Affiliation(s)
| | - Jennifer Y Chen
- Department of Medicine
- The Liver Center, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
32
|
Rudbaek JJ, Agrawal M, Torres J, Mehandru S, Colombel JF, Jess T. Deciphering the different phases of preclinical inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2024; 21:86-100. [PMID: 37950021 PMCID: PMC11148654 DOI: 10.1038/s41575-023-00854-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/12/2023]
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory disease (IMID) of the gastrointestinal tract and includes two subtypes: Crohn's disease and ulcerative colitis. It is well-recognized that IBD is associated with a complex multifactorial aetiology that includes genetic predisposition and environmental exposures, with downstream dysregulation of systemic immune function and host-microbial interactions in the local environment in the gut. Evidence to support the notion of a multistage development of IBD is growing, as has been observed in other IMIDs such as rheumatoid arthritis and systemic lupus erythematosus. With the rising worldwide incidence of IBD, it is increasingly important to understand the complex interplay of pathological events during the different stages of disease development to enable IBD prediction and prevention strategies. In this article, we review comprehensively the current evidence pertaining to the preclinical phase of IBD, including at-risk, initiation and expansion phases. We also discuss the framework of preclinical IBD, expanding on underlying pathways in IBD development, future research directions and IBD development in the context of other IMIDs.
Collapse
Affiliation(s)
- Jonas J Rudbaek
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Section for Biomarkers, Immunology and Antibodies, Department for Congenital Disorders, Statens Serum Institut, Copenhangen, Denmark
| | - Manasi Agrawal
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joana Torres
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
- Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Saurabh Mehandru
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark.
- Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
33
|
Marafini I, Laudisi F, Salvatori S, Lavigna D, Venuto C, Giannarelli D, Monteleone G. Diagnostic value of anti-integrin αvβ6 antibodies in ulcerative colitis. Dig Liver Dis 2024; 56:55-60. [PMID: 37407314 DOI: 10.1016/j.dld.2023.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
Ulcerative colitis (UC)-related mucosal inflammation is characterized by the production of various autoantibodies with limited clinical relevance. Recent studies have shown that circulating levels of IgG against integrin αvβ6 are increased in UC patients as compared to Crohn's disease (CD) patients and healthy controls (HC). The present study assessed the diagnostic value of circulating IgG anti-αvβ6 in UC. Sera were prospectively collected from 108 outpatients with UC, 103 patients with CD, and 62 HC, and the levels of IgG anti-αvβ6 were measured using a commercially available ELISA kit. The cut-off for positive results was defined as the 95th percentile of the values of the autoantibodies in HC serum samples. Levels of IgG anti-αvβ6 were significantly higher in UC than in CD patients, including those with colonic localization, and HC. Fifty-six of the 108 (51.8%) UC patients had a positive test whereas only 17/103 (16.5%) patients with CD, and among these, 4/16 (25%) patients with colonic CD, were positive. In UC, there was no statistical difference between patients with IgG anti-αvβ6 positivity and those negative in terms of clinical disease activity, fecal calprotectin values, and disease extent. The sensitivity, specificity, predictive positive value, and predictive negative value of the test to differentiate between UC and CD were 51.9% (C.I.42.4-61.3), 83.5% (C.I. 76.3-90.7), 76.7% (C.I. 67.0-86.4), and 62.3% (C.I. 54.2-70.4) respectively. Our study confirms that anti-αvβ6 antibodies are demonstrable in the serum of the majority of UC patients and suggests the necessity of further research to understand if the anti-αvβ6 antibody determination could have a place in the clinical decision-making of IBD.
Collapse
Affiliation(s)
- Irene Marafini
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Salvatori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Diletta Lavigna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Chiara Venuto
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
34
|
Andalucía C, Martínez-Prat L, Bentow C, Aure MA, Horn MP, Mahler M. Clinical Validity of Anti-Proteinase 3 Antibodies in Patients with Inflammatory Bowel Disease: A Short Meta-Analysis. Diagnostics (Basel) 2023; 13:3682. [PMID: 38132266 PMCID: PMC10742424 DOI: 10.3390/diagnostics13243682] [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: 11/06/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Anti-neutrophil cytoplasmic antibodies (ANCA) directed to proteinase 3 (PR3) represent highly established markers for patients with ANCA-associated vasculitis (AAV). PR3-ANCA have also demonstrated utility in the management of inflammatory bowel disease (IBD). More specifically, PR3-ANCA discriminate individuals with ulcerative colitis (UC) from Crohn's disease (CD) patients and are associated with disease severity, activity, and treatment non-response. Here, we aim to summarize the current data on the diagnostic utility of PR3-ANCA in IBD. A structured, systematic literature review, including three electronic databases, was conducted on June 6th, 2023, to identify studies assessing the diagnostic accuracy of the QUANTA Flash® PR3 assay in UC vs. CD patients. Electronic searches were supplemented by hand searching. A hierarchical, bivariate, mixed-effect meta-analysis was conducted using the metandi function, as per the Cochrane collaboration recommendations. Study quality was assessed using the QUADAS-2 tool, which considers the risk of bias and applicability. Six out of a hundred and eleven citations met the inclusion criteria and reported QUANTA Flash® PR3 diagnostic accuracy in UC vs. CD (UC, n = 667, CD, n = 682 patients). The sensitivity/specificity point estimate for UC was 34.9%/95.9%. This resulted in a Diagnostic Odds Ratio (DOR) of 12.6. The risk of bias was low in the index test and reference standard domains. Four of the six studies (67%) showed an unclear risk of bias in patient selection and in flow and timing domains. All studies had low concerns about applicability in all the domains. PR3-ANCA measured with the QUANTA Flash® PR3 assay represent novel diagnostic markers in IBD and enables discrimination between UC and CD.
Collapse
Affiliation(s)
- Carmen Andalucía
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA; (C.A.); (L.M.-P.); (C.B.); (M.A.A.)
| | - Laura Martínez-Prat
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA; (C.A.); (L.M.-P.); (C.B.); (M.A.A.)
| | - Chelsea Bentow
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA; (C.A.); (L.M.-P.); (C.B.); (M.A.A.)
| | - Mary Ann Aure
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA; (C.A.); (L.M.-P.); (C.B.); (M.A.A.)
| | - Michael P. Horn
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
| | - Michael Mahler
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA; (C.A.); (L.M.-P.); (C.B.); (M.A.A.)
| |
Collapse
|
35
|
Chen Y, Lan C, Zhong W, Song K, Ma Z, Huang L, Zhu Y, Xia H. Plasma anti-myosin autoantibodies in the diagnosis of necrotizing enterocolitis. Eur J Pediatr 2023; 182:5203-5210. [PMID: 37715022 PMCID: PMC10640473 DOI: 10.1007/s00431-023-05188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/17/2023]
Abstract
We aimed to assess whether autoantibodies can be used as biomarkers for necrotizing enterocolitis (NEC) and applied for its early diagnosis. A prospective observational study was conducted in neonates with suspected NEC abdominal distension (the developmental study), which consisted of 50 neonates finally divided into NEC (n = 24) and non-NEC (n = 26) cohorts based on follow-up results. Serum samples were collected within 48 h of illness onset and used for screening NEC-associated plasma autoantibodies by autoantigen microarray. Additionally, we validated anti-myosin autoantibodies by enzyme-linked immunosorbent assay (ELISA) in an independent validation study, for which we selected plasma samples within 48 h of onset of NEC (n = 38) and samples of gestational age- and weight-matched controls (n = 13). Autoantigen microarray revealed that both IgG and IgM anti-myosin autoantibodies in plasma from neonates with NEC were significantly higher than those in neonates with other diagnoses. ELISA showed that plasma anti-myosin autoantibodies increased in the NEC cohort, with 1.5-fold higher levels than in the non-NEC cohort. Anti-myosin autoantibodies were able to distinguish NEC from non-NEC, achieving an area under the curve (AUC) of 0.8856 (95% confidence interval (CI): 0.7918-0.9795), with sensitivity of 81.58% and specificity of 76.93%. Plasma anti-myosin autoantibodies were significantly higher in all three subtypes of NEC (P < 0.0001 for NEC I; P = 0.0018 for NEC II; P = 0.0011 for NEC III), especially in NEC stage I than that in the non-NEC controls. CONCLUSION Anti-myosin autoantibodies may be applied as a promising diagnostic marker for NEC, especially for NEC stage I. WHAT IS KNOWN • Intestinal damage and self-antigen exposure may lead to increased autoantibodies, and they are widely used as biomarkers for diagnosing inflammatory bowel disease. • Necrotizing enterocolitis (NEC) is a devastating disease with overwhelming inflammation and immune dysregulation. WHAT IS NEW • Increased autoantibodies were present in patients with NEC, even before typical X-ray manifestations. • Anti-myosin autoantibodies may be applied as a promising diagnostic marker for NEC.
Collapse
Affiliation(s)
- Yuqiong Chen
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, CN 510630, China
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Chaoting Lan
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Weiyong Zhong
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Kai Song
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Zuyi Ma
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Lihua Huang
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| | - Yun Zhu
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| | - Huimin Xia
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, CN 510630, China.
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| |
Collapse
|
36
|
Nowak JK, Kalla R, Satsangi J. Current and emerging biomarkers for ulcerative colitis. Expert Rev Mol Diagn 2023; 23:1107-1119. [PMID: 37933807 DOI: 10.1080/14737159.2023.2279611] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Ulcerative colitis (UC) is a chronic illness requiring lifelong management that could be enhanced by personalizing care using biomarkers. AREAS COVERED The main biomarker discovery modalities are reviewed, highlighting recent results across the spectrum of applications, including diagnostics (serum anti-αvβ6 antibodies achieving an area under the curve [AUC] = 0.99; serum oncostatin M AUC = 0.94), disease activity assessment (fecal calprotectin and serum trefoil factor 3: AUC > 0.90), prognostication of the need for treatment escalation (whole blood transcriptomic panels and CLEC5A/CDH2 ratio: AUC > 0.90), prediction of treatment response, and early identification of patients with subclinical disease. The use of established biomarkers is discussed, along with new evidence regarding autoantibodies, proteins, proteomic panels, transcriptomic signatures, deoxyribonucleic acid methylation patterns, and UC-specific glycomic and metabolic disturbances. EXPERT OPINION Novel biomarkers will pave the way for optimized UC care. However, validation, simplification, and direct clinical translation of complex models may prove challenging. Currently, few candidates exist to assess key characteristics, such as UC susceptibility, histological disease activity, drug response, and long-term disease behavior. Further research will likely not only reveal new tools to tackle these issues but also contribute to understanding UC pathogenesis mechanisms.
Collapse
Affiliation(s)
- Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Rahul Kalla
- Medical Research Council Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Experimental Medicine Division, University of Oxford, Oxford, UK
| |
Collapse
|
37
|
Rodríguez-Lago I, Blackwell J, Mateos B, Marigorta UM, Barreiro-de Acosta M, Pollok R. Recent Advances and Potential Multi-Omics Approaches in the Early Phases of Inflammatory Bowel Disease. J Clin Med 2023; 12:jcm12103418. [PMID: 37240524 DOI: 10.3390/jcm12103418] [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: 03/20/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammatory bowel disease leads to debilitating gastrointestinal symptoms and reduced quality of life, resulting in a significant burden on healthcare utilization and costs. Despite substantial advancements in diagnosis and treatment, there may still be considerable delays in diagnosing some patients. To reduce disease progression before the full disease spectrum appears and improve prognostic outcomes, several strategies have concentrated on early intervention and prevention. Recent evidence shows that initial immune response changes and endoscopic lesions may exist for years before diagnosis, implying the existence of a preclinical phase of inflammatory bowel disease comparable to findings in other immune-mediated disorders. In this review, we highlight the most relevant findings regarding preclinical inflammatory bowel disease and the prospective role of novel omics techniques in this field.
Collapse
Affiliation(s)
- Iago Rodríguez-Lago
- Gastroenterology Department, Hospital Universitario de Galdakao, 48960 Galdakao, Spain
- Biocruces Bizkaia Health Research Institute, 48960 Galdakao, Spain
- Deusto University, 48007 Bilbao, Spain
| | | | - Beatriz Mateos
- Integrative Genomics Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Urko M Marigorta
- Integrative Genomics Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Sciences, 48009 Bilbao, Spain
| | - Manuel Barreiro-de Acosta
- Gastroenterology Department, Hospital Clínico Universitario de Santiago, 15706 Santiago de Compostela, Spain
| | - Richard Pollok
- Gastroenterology Department, St George's University of London, London SW17 0RE, UK
| |
Collapse
|