1
|
Loh L, Orlicky DJ, Spengler A, Domenico J, Klarquist J, Levens C, Celli S, Kofonow JM, Robertson CE, Lantz O, Legoux F, Frank DN, Matsuda J, Norman PJ, Kuhn K, Onyiah J, Gapin L. MAIT cells exacerbate colonic inflammation in a genetically diverse murine model of spontaneous colitis. Mucosal Immunol 2025:S1933-0219(25)00053-4. [PMID: 40425090 DOI: 10.1016/j.mucimm.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 05/01/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
IL-17-producing lymphocytes are involved in both tissue repair and the propagation of inflammation, with their effects highly context-dependent. Mucosal-Associated-Invariant-T-cells (MAIT), a subset of innate-like T cells with features of both Th1 and Th17 lineages, are increasingly recognized for their roles in mucosal immunity. Here, we identified the Collaborative-Cross CC011/Unc strain, which spontaneously develops chronic colitis, as being enriched for MAIT cells. This expansion coincides with an age-related loss of intestinal barrier permeability and colonic inflammation. Microbiota from CC011 mice activated MAIT cells in an MR1-dependent manner and selectively promoted the accumulation of MAIT17 cells in peripheral tissues. Single-cell transcriptomic analyses revealed colon MAIT cells from colitic CC011 mice expressed a pathogenic Th17-like signature, characterized by IL-1 and IL-23 signaling, IL-17A and IFNγ co-expression, and upregulation of IL-23R, features that correlated with inflammatory Ly6Chi monocyte abundance. Genetic deletion of Traj33, essential for MAIT development, significantly reduced colonic inflammation in this model. These findings demonstrate that MAIT cells integrate microbial and cytokine cues to adopt a pathogenic effector phenotype that exacerbates chronic intestinal inflammation.
Collapse
Affiliation(s)
- Liyen Loh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| | - David J Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andrea Spengler
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joanne Domenico
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Cassandra Levens
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sofia Celli
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer M Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Charles E Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Olivier Lantz
- Institut Curie, Paris Sciences et Lettres University, Paris, France
| | - Francois Legoux
- Institut Curie, Paris Sciences et Lettres University, Paris, France
| | - Daniel N Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer Matsuda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA; Mouse Genetics Core, National Jewish Health, Denver, CO, USA
| | - Paul J Norman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA; Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kristine Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joseph Onyiah
- Division of Gastroenterology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
2
|
Munasinghe S, Chandrakasan S. Maternal Microchimerism, a Red Flag or a Red Herring in Very-Early-Onset Inflammatory Bowel Disease? Inflamm Bowel Dis 2025:izaf070. [PMID: 40349188 DOI: 10.1093/ibd/izaf070] [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: 03/08/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Sachith Munasinghe
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shanmuganathan Chandrakasan
- Department of Pediatrics, Children's Health Care of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Division of Hematology, Oncology and BMT, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
3
|
Chen Y, He X, Li D, Qiao L, Lu G. Secondary hemophagocytic lymphohistiocytosis in a 5-month-old infant with IBD post-COVID-19: a case report. Front Pediatr 2025; 13:1536066. [PMID: 40376626 PMCID: PMC12078334 DOI: 10.3389/fped.2025.1536066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/15/2025] [Indexed: 05/18/2025] Open
Abstract
Background COVID-19 is known to induce cytokine storms and inappropriate cytotoxic immune responses. Hemophagocytic lymphohistiocytosis (HLH) is an underrecognized condition due to a hyperinflammatory syndrome characterized by fulminant hypercytokinemia with a high mortality burden. Cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induced secondary HLH during and post-infection have been sparsely reported in children. Inflammatory bowel disease (IBD) is a chronic, relapsing disorder of the gastrointestinal tract with a multifactorial etiology involving genetic, environmental, and immunological factors. To date, secondary HLH associated with COVID-19 in very early-onset inflammatory bowel disease (VEO-IBD) has not been reported. This case report aims to enhance understanding of the clinical manifestations of VEO-IBD and HLH, thereby facilitating the timely diagnosis and management of this rare condition. Case presentation We present the case of a 5-month-old Chinese female infant diagnosed with HLH following COVID-19 infection. The patient presented with hemophagocytic syndrome, which included recurrent fever, hepatosplenomegaly, cytopenia, hyperferritinemia, hypertriglyceridemia, and hypofibrinogenemia, after exposure to her mother, who had been diagnosed with COVID-19. Whole-exome sequencing(WES) identified heterozygous mutations in the IL-10RA gene: c.537 G > A (inherited from her mother) and c.301 C > T (inherited from her father), who was ultimately identified as having VEO-IBD. Despite receiving nutritional support, intravenous immunoglobulin (IVIG), and dexamethasone therapy, the patient continued to experience anemia, diarrhea, and refractory gastrointestinal bleeding. Following a brief improvement after interventional treatment, the parents declined further medical interventions, signed for discharge, and the infant sadly passed away three months later. Conclusion Rare genetic variants play a pivotal role in the pathogenesis of VEO-IBD, particularly in infants diagnosed before the age of one. These cases often demonstrate resistance to various immunosuppressive therapies and have a poor prognosis with conventional treatments. Our findings highlight the potential increased risk of severe HLH in patients with VEO-IBD and concurrent COVID-19, underscoring the need for comprehensive and vigilant differential diagnosis when patients exhibit symptoms suggestive of multi-organ damage.
Collapse
Affiliation(s)
| | | | | | - Lina Qiao
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children’s Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guoyan Lu
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children’s Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Arienzo F, Giovannoni I, Diamanti A, Trovato CM, De Angelis P, Imondi C, Alaggio R, Francalanci P. Paediatric Congenital Enteropathies: Clinical and Histological Review. Diagnostics (Basel) 2025; 15:946. [PMID: 40310351 PMCID: PMC12025760 DOI: 10.3390/diagnostics15080946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Paediatric congenital enteropathies (PCEs) are a group of rare inherited diseases with a typical early onset in life. Prompt identification and treatment are crucial to avoid potentially fatal consequences. This review aims to provide a paradigmatic framework for clinical and histological identification of PCEs, with an emphasis on congenital conditions involving epithelial shape, trafficking and polarity, enteroendocrine function, immunomodulatory diseases, and extremely early onset inflammatory bowel illness. A proper classification is founded on histopathological characteristics and clinical parameters (such as consanguinity, anomalies in amniotic fluid, prenatal expression or early neonatal onset, stool appearance, persistence of symptoms despite fasting, and extra-intestinal manifestations, etc.). The increasing accessibility and convenience of genetic tests has also accelerated the identification of genes related to specific phenotypes of PCEs, improving the diagnostic and care pathway. As a "niche" pathology, PCEs are susceptible to misdiagnosis due to a limited awareness of these entities, and their identification requires extensive training and specialized facilities. The aim of our review is to emphasize the importance of an integrated approach, combining clinical, histological, and molecular analysis, to achieve a definitive diagnosis and guide the treatment.
Collapse
Affiliation(s)
- Francesca Arienzo
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | | | - Antonella Diamanti
- Hepatology, Gastroenterology and Nutrition Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Chiara Maria Trovato
- Hepatology, Gastroenterology and Nutrition Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Paola De Angelis
- Hepatology, Gastroenterology and Nutrition Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Chiara Imondi
- Hepatology, Gastroenterology and Nutrition Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Paola Francalanci
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
5
|
Andrews AR, Putra J. Special Considerations in Pediatric Inflammatory Bowel Disease Pathology. Diagnostics (Basel) 2025; 15:831. [PMID: 40218181 PMCID: PMC11988757 DOI: 10.3390/diagnostics15070831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/14/2025] Open
Abstract
Inflammatory bowel disease (IBD) in the pediatric population presents distinct characteristics compared to adult cases. Pathology plays a critical role in its diagnosis, and this review underscores key considerations in the pathologic evaluation of pediatric IBD. Recognizing inflammatory patterns in the upper gastrointestinal tract can improve disease classification and aid in diagnosing IBD in certain scenarios, such as isolated upper gastrointestinal or small bowel involvement. Additionally, familiarity with distinctive subtypes, including IBD associated with primary sclerosing cholangitis and monogenic forms of IBD, supports early comorbidity detection, enhances patient management, and improves prognostication.
Collapse
Affiliation(s)
- Alicia R. Andrews
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| | - Juan Putra
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Noble A, Adams A, Nowak J, Cheng G, Nayak K, Quinn A, Kristiansen M, Kalla R, Ventham NT, Giachero F, Jayamanne C, Hansen R, Hold GL, El-Omar E, Croft NM, Wilson D, Beattie RM, Ashton JJ, Zilbauer M, Ennis S, Uhlig HH, Satsangi J. The Circulating Methylome in Childhood-Onset Inflammatory Bowel Disease. J Crohns Colitis 2025; 19:jjae157. [PMID: 39365013 PMCID: PMC11945304 DOI: 10.1093/ecco-jcc/jjae157] [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: 04/08/2024] [Revised: 07/16/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND The genetic contribution to inflammatory bowel disease (IBD), encompassing both Crohn's disease (CD) and ulcerative colitis (UC), accounts for around 20% of disease variance, highlighting the need to characterize environmental and epigenetic influences. Recently, considerable progress has been made in characterizing the adult methylome in epigenome-wide association studies. METHODS We report detailed analysis of the circulating methylome in 86 patients with childhood-onset CD and UC and 30 controls using the Illumina Infinium Human MethylationEPIC platform. RESULTS We derived and validated a 4-probe methylation biomarker (RPS6KA2, VMP1, CFI, and ARHGEF3), with specificity and high diagnostic accuracy for pediatric IBD in UK and North American cohorts (area under the curve: 0.90-0.94). Significant epigenetic age acceleration is present at diagnosis, with the greatest observed in CD patients. Cis-methylation quantitative trait loci (meQTL) analysis identifies genetic determinants underlying epigenetic alterations notably within the HLA 6p22.1-p21.33 region. Passive smoking exposure is associated with the development of UC rather than CD, contrary to previous findings. CONCLUSIONS These data provide new insights into epigenetic alterations in IBD and illustrate the reproducibility and translational potential of epigenome-wide association studies in complex diseases.
Collapse
Affiliation(s)
- Alexandra Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| | - Alex Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jan Nowak
- Department of Paediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Guo Cheng
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - Komal Nayak
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Aisling Quinn
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| | - Mark Kristiansen
- UCL Genomics, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Rahul Kalla
- Medical Research Council Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nicholas T Ventham
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Federica Giachero
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke’s Hospital, Cambridge, UK
| | - Chamara Jayamanne
- Department of Paediatrics, John Radcliffe Hospital, Oxford University Hospital NHS Trust, Oxford, UK
| | - Richard Hansen
- Department of Child Health, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Georgina L Hold
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
| | - Emad El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicholas M Croft
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David Wilson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
- Department of Child Life and Health, University of Edinburgh, Edinburgh, UK
| | - R Mark Beattie
- Department of Paediatric Gastroenterology, Southampton Children’s Hospital, Southampton, UK
| | - James J Ashton
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
- Department of Paediatric Gastroenterology, Southampton Children’s Hospital, Southampton, UK
| | - Matthias Zilbauer
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke’s Hospital, Cambridge, UK
| | - Sarah Ennis
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Jones K, Plotkin L, Loukogeorgakis S, Cytter-Kuint R, Worth A, Turner D. The Management of Internal Fistulizing Crohn's Disease in a Child: More Than Meets the Eye. Gastroenterology 2025; 168:471-479.e1. [PMID: 39477027 DOI: 10.1053/j.gastro.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024]
Affiliation(s)
- Kelsey Jones
- Paediatric Gastroenterology, Great Ormand Street Hospital, London, United Kingdom
| | - Luba Plotkin
- Juliet Keidan Institute of Pediatric Gastroenterology, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stavros Loukogeorgakis
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, Research and Teaching Department of Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ruth Cytter-Kuint
- Pediatric Radiology Unit, Radiology Department, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Austen Worth
- Department of Pediatric Immunology and Gene Therapy, Great Ormand Street Hospital, London, United Kingdom
| | - Dan Turner
- Paediatric Gastroenterology, Great Ormand Street Hospital, London, United Kingdom; Juliet Keidan Institute of Pediatric Gastroenterology, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
8
|
Ferro J, Francalanci P, Angerilli V, Cafferata B, D'Armiento M, Buccoliero AM, Franca RA, Vanoli A, Macciomei MC, Santoro L, Alaggio R, Fassan M, Mastracci L, Sacchi D, Giordano C, Pilozzi E, Giustiniani MC, Panarese I, Grillo F, Parente P. Histologic alterations are common in Monogenic Disease patients with 'healthy' endoscopy: Results from a GIPAD-GIPPI multicenter study. Hum Pathol 2025; 157:105763. [PMID: 40132689 DOI: 10.1016/j.humpath.2025.105763] [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: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVE Very early-onset inflammatory bowel disease (VEO-IBD) is a clinical umbrella term referring to IBD-like symptoms arising in children before 6 years of age, encompassing 'pure' IBD (Crohn's Colitis/Ulcerative Colitis)/non IBD colitis, and monogenic diseases (MDs), the latter often related to primary immunodeficiency disorders. A multidisciplinary approach is imperative for correct therapeutic management, as endoscopy and histology are not always completely informative. In this setting, the study aims to describe the extent/features of histologic lesions in both endoscopically damaged mucosa and otherwise endoscopically healthy (normal/near normal) mucosa. METHODS Endoscopic data were retrospectively recorded, and histologic slides were collegially re-evaluated in a 93 VEO-IBD multicenter cohort, 76 (76/93 - 81,7 %) of which with complete endoscopic/histologic data. RESULTS At endoscopy, lesions were reported by the clinician in 66/76 (86,8 %) cases. When endoscopic lesions were reported, histologic damage was also seen. Interestingly, histologic mucosal damage was also documented in 43,3 % (13/30) of cases with endoscopically healthy/nearly healthy mucosa. This misalignment between endoscopy and pathology was seen in about a third of (29,1 % - 7/24) 'true' IBD and all MDs (100 % - 6/6) (p = 0.0029). CONCLUSION In VEO-IBD, histologic lesions can be present in endoscopically 'healthy' intestinal mucosa. This finding is more frequent in MDs, suggesting the need to accurately sample all the mucosal tract in VEO-IBD patients, even when no endoscopic lesions are seen at endoscopy.
Collapse
Affiliation(s)
- Jacopo Ferro
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini, 5, 16147, Genova, Italy.
| | - Paola Francalanci
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Piazza Sant'Onofrio 4, 00165, Roma, Italy.
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Via Ospedale Vecchio, 35128, Padua, Italy.
| | - Barbara Cafferata
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini, 5, 16147, Genova, Italy.
| | - Maria D'Armiento
- Pathology Unit, Department of Public Health, University of Naples Federico II, Via Sergio Pansini, 80131, Napoli, Italy.
| | - Anna Maria Buccoliero
- Pathology Unit Meyer Children's Hospital IRCCS, Viale Gaetano Pieraccini 24, 50100, Firenze, Italy.
| | - Raduan Ahmed Franca
- Department of Pathology, Santobono-Pausilipon, Children's Hospital, via Ravaschieri 8, 80122, Naples, Italy.
| | - Alessandro Vanoli
- Department of Molecular Medicine, University of Pavia, Strada Nuova 65, 27100, Pavia, Italy; Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, Viale Golgi 19, 27100, Pavia, Italy.
| | - Maria Cristina Macciomei
- Pathology Unit, Azienda Ospedaliera San Camillo-Forlanini, Circonvallazione Gianicolense 87, 00152, Roma, Italy.
| | - Luisa Santoro
- Pathology Unit, Azienda Ospedaliera Padova, Via Ospedale Vecchio, 35128, Padova, Italy.
| | - Rita Alaggio
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Piazza Sant'Onofrio 4, 00165, Roma, Italy.
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Via Ospedale Vecchio, 35128, Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Viale Gattamelata, 35128, Padua, Italy.
| | - Luca Mastracci
- Anatomic Pathology Unit IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy; Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Via Balbi 5, 16126, Genoa, Italy.
| | - Diana Sacchi
- Pathology Unit, Ospedale Ca' Foncello, Piazzale dell'Ospedale 1, 31100, Treviso, Italy.
| | - Carla Giordano
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza, University of Rome, Unit of Anatomic Pathology Morphologic and Molecular Sant'Andrea Hospital, via di Grottarossa 1035, 00189, Rome, Italy.
| | - Maria Cristina Giustiniani
- Department of Pathology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Gemelli 8, 00165, Rome, Italy.
| | - Iacopo Panarese
- Pathology Unit, Università Vanvitelli di Napoli, via Costantinopoli, 80138, Napoli, Italy.
| | - Federica Grillo
- Anatomic Pathology Unit IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy; Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Via Balbi 5, 16126, Genoa, Italy.
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
9
|
Dogan S, Selen R, Ozbay Hosnut F, Ozdel S, Dogu F, Ikinciogullari A, Aytekin C. Successful Use of Anakinra in a Patient with IL-10R Beta Deficiency: A Case Report. PEDIATRIC ALLERGY, IMMUNOLOGY, AND PULMONOLOGY 2025; 38:32-35. [PMID: 39950990 DOI: 10.1089/ped.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Background: Interleukin-10 (IL-10) is a crucial anti-inflammatory cytokine essential for maintaining immune homeostasis, particularly in the gastrointestinal system. Defects in the IL-10 signaling pathway, including mutations in interlaukin-10 receptor (IL-10R) beta, have been implicated in early-onset inflammatory bowel disease (IBD), often resistant to conventional treatments. Case Presentation: We report a 1.5-month-old male patient with IL-10R beta deficiency presenting with fever, vomiting, malnutrition, and sepsis. Despite normal initial evaluations, persistent fever and elevated inflammatory markers prompted the initiation of anakinra, an interleukin-1 receptor antagonist. Genetic testing confirmed a homozygous deletion in the IL10RB gene. Anakinra led to significant clinical improvement, including weight gain and symptom resolution. The patient was enrolled in an allogeneic hematopoietic stem cell transplantation (HSCT) program and successfully received HSCT from an HLA-matched related donor. Discussion: IL-10R beta deficiency presents with severe and early-onset symptoms, often unresponsive to standard IBD therapies. Anakinra has shown promise in bridging to HSCT by reducing inflammation and improving clinical outcomes in patients with IL-10 pathway defects. This case highlights the effectiveness of anakinra as a treatment strategy in severe, refractory IBD associated with IL-10R beta deficiency and underscores the importance of genetic testing for accurate diagnosis and treatment planning. Conclusion: Anakinra may provide significant clinical benefits in patients with IL-10R beta deficiency, serving as a bridge to definitive treatment with HSCT. Early genetic diagnosis and targeted therapy are crucial for managing this challenging condition.
Collapse
Affiliation(s)
- Selcuk Dogan
- Department of Pediatric Immunology and Allergy, Dr. Sami Ulus Children's Health and Diseases Training and Research Hospital, Ankara, Turkiye
| | - Ridvan Selen
- Department of Pediatric Immunology and Allergy, Dr. Sami Ulus Children's Health and Diseases Training and Research Hospital, Ankara, Turkiye
| | - Ferda Ozbay Hosnut
- Department of Pediatric Gastroenterology, Dr. Sami Ulus Children's Health and Diseases Training and Research Hospital, Ankara, Turkiye
| | - Semanur Ozdel
- Department of Pediatric Rheumatology, Dr. Sami Ulus Children's Health and Diseases Training and Research Hospital, Ankara, Turkiye
| | - Figen Dogu
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Aydan Ikinciogullari
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Caner Aytekin
- Department of Pediatric Immunology and Allergy, Dr. Sami Ulus Children's Health and Diseases Training and Research Hospital, Ankara, Turkiye
| |
Collapse
|
10
|
Li J, Jacobse J, Pilat JM, Kaur H, Gu W, Kang SW, Rusznak M, Huang HI, Barrera J, Oloo PA, Roland JT, Hawkins CV, Pahnke AP, Khalil M, Washington MK, Wilson KT, Williams CS, Peebles RS, Konnikova L, Choksi YA, Hammer GE, Lau KS, Goettel JA. Interleukin-10 production by innate lymphoid cells restricts intestinal inflammation in mice. Mucosal Immunol 2025:S1933-0219(25)00023-6. [PMID: 39988202 DOI: 10.1016/j.mucimm.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Interleukin-10 (IL-10) is an immunomodulatory cytokine critical for intestinal immune homeostasis. IL-10 is produced by various immune cells but IL-10 receptor signaling in intestinal CX3CR1+ mononuclear phagocytes is necessary to prevent spontaneous colitis in mice. Here, we utilized fluorescent protein reporters and cell-specific targeting and found that Rorc-expressing innate lymphoid cells (ILCs) produce IL-10 in response to anti-CD40-mediated intestinal inflammation. Deletion of Il10 specifically in Rorc-expressing ILCs led to phenotypic changes in intestinal macrophages and exacerbated both innate and adaptive immune-mediated models of experimental colitis. The population of IL-10+ producing ILCs shared markers with both ILC2 and ILC3 with nearly all ILC3s being of the NCR+ subtype. Interestingly, Ccl26 was enriched in IL-10+ ILCs and was markedly reduced in IL-10-deficient ILC3s. Since CCL26 is a ligand for CX3CR1, we employed RNA in situ hybridization and observed increased numbers of ILCs in close proximity to Cx3cr1-expressing cells under inflammatory conditions. Finally, we generated transgenic RorctdTomato reporter mice that faithfully marked RORγt+ cells that could rescue disease pathology and aberrant macrophage phenotype following adoptive transfer into mice with selective Il10 deficiency in ILC3s. These results demonstrate that IL-10 production by a population of ILCs functions to promote immune homeostasis in the intestine possibly via direct effects on intestinal macrophages.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212
| | - Jennifer M Pilat
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harsimran Kaur
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Weihong Gu
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Seung Woo Kang
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark Rusznak
- Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hsin-I Huang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Julio Barrera
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Pauline A Oloo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph T Roland
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V Hawkins
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew P Pahnke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marian Khalil
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA; Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Obstetrics Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yash A Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gianna Elena Hammer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ken S Lau
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Computational Systems Biology, Vanderbilt University, Nashville, TN, USA
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
11
|
Calvez V, Puca P, Di Vincenzo F, Del Gaudio A, Bartocci B, Murgiano M, Iaccarino J, Parand E, Napolitano D, Pugliese D, Gasbarrini A, Scaldaferri F. Novel Insights into the Pathogenesis of Inflammatory Bowel Diseases. Biomedicines 2025; 13:305. [PMID: 40002718 PMCID: PMC11853239 DOI: 10.3390/biomedicines13020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs), encompassing Crohn's disease and ulcerative colitis, are complex chronic disorders characterized by an intricate interplay between genetic predisposition, immune dysregulation, gut microbiota alterations, and environmental exposures. This review aims to synthesize recent advances in IBD pathogenesis, exploring key mechanisms and potential avenues for prevention and personalized therapy. A comprehensive literature search was conducted across major bibliographic databases, selecting the most recent and impactful studies on IBD pathogenesis. The review integrates findings from multi-omics analyses, single-cell transcriptomics, and longitudinal cohort studies, focusing on immune regulation, gut microbiota dynamics, and environmental factors influencing disease onset and progression. Immune dysregulation, including macrophage polarization (M1 vs. M2) and Th17 activation, emerges as a cornerstone of IBD pathogenesis. Dysbiosis, as a result of reduced alpha and beta diversity and overgrowth of harmful taxa, is one of the main contributing factors in causing inflammation in IBD. Environmental factors, including air and water pollutants, maternal smoking, and antibiotic exposure during pregnancy and infancy, significantly modulate IBD risk through epigenetic and microbiota-mediated mechanisms. While recent advances have supported the development of new therapeutic strategies, deeply understanding the complex dynamics of IBD pathogenesis remains challenging. Future efforts should aim to reduce the burden of disease with precise, personalized treatments and lower the incidence of IBD through early-life prevention and targeted interventions addressing modifiable risk factors.
Collapse
Affiliation(s)
- Valentin Calvez
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Pierluigi Puca
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Federica Di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Jacopo Iaccarino
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Erfan Parand
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Daniele Napolitano
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
| | - Daniela Pugliese
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Franco Scaldaferri
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| |
Collapse
|
12
|
Xu X, Zhou Y, Tan Z, Huang Y, Dong K, Gu Y, Chen J, Yu Z. Risk factors for stoma and incision complications of enterostomy in children with very early-onset inflammatory bowel disease: a prospective cohort study. Eur J Pediatr 2025; 184:146. [PMID: 39828783 DOI: 10.1007/s00431-024-05952-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Enterostomy is utilized to mitigate severe clinical symptoms in children with very early-onset inflammatory bowel disease (VEO-IBD) and to provide a window for stem cell transplantation. Nevertheless, the incidence of postoperative complications is significant, and there is currently a lack of research exploring the risk factors associated with complications related to the stoma and incision following the procedure. The objective of this study is to investigate the risk factors for stoma and incision complications after enterostomy in patients with VEO-IBD. From January 2015 to December 2023, 49 children with VEO-IBD who underwent enterostomy were enrolled in the study. Demographic characteristics, blood biochemical indices, weighted Pediatric Crohn's Disease Activity Index (wPCDAI), and enterostomy-related information were prospectively collected. Multivariate logistic regression was employed to identify the risk factors for ostomy and incision-related complications. All 49 included VEO-IBD children had interleukin-10 (IL-10) signaling defects, with 27 (55.1%) having stomal-related complications and 10 (20.4%) had incision complications after enterostomy. Univariate analysis revealed that wPCDAI (OR, 1.03; 95% CI, 1.00-1.07; P = 0.05) showed a tendency towards statistical significance in the occurrence of ostomy complications. Weight-for-age Z-score (WAZ) (OR, 0.57; 95% CI, 0.39-0.84; P = 0.004), height-for-age Z-score (HAZ) (OR, 0.57; 95% CI, 0.37-0.88; P = 0.01), type of surgery (OR, 0.12; 95% CI, 0.03-0.56, P = 0.007), C-reactive protein (CRP) (OR, 1.02; 95% CI, 1.01-1.04; P = 0.007), and wPCDAI (OR, 1.08; 95% CI, 1.01-1.14; P = 0.009) demonstrated statistical significance in the occurrence of incision complications. However, multivariate binary logistic regression did not reveal any statistically significant factors. CONCLUSION Although emergency surgery is unavoidable, our study suggests that improving nutritional status, reducing CRP levels, and increasing preoperative wPCDAI scores may help reduce post-enterostomy stoma and incision complications in VEO-IBD children with interleukin-10 (IL-10) signaling defects. Further large-scale studies are needed to confirm these findings. WHAT IS KNOWN • Enterostomy is commonly used to manage severe symptoms in children with VEO-IBD and to provide a window for stem cell transplantation. • The incidence of postoperative complications, including stoma and incision-related issues, is significant in these patients. WHAT IS NEW • This study identifies potential risk factors for stoma and incision complications following enterostomy in children with VEO-IBD, particularly those with IL-10 signaling defects. • Factors such as nutritional status (WAZ and HAZ), CRP levels, type of surgery, and the wPCDAI were found to be associated with stoma and incision complications in univariate analysis, although multivariate analysis did not show statistical significance for these factors.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Department of Gastroenterology, Children's Hospital of Fudan University, No. 399, Wanyuan Rd., Minhang District, Shanghai, China.
| | - Yiwen Zhou
- Department of Gastroenterology, Children's Hospital of Fudan University, No. 399, Wanyuan Rd., Minhang District, Shanghai, China
| | - Zhixin Tan
- School of Nursing, Fudan University, Shanghai, China
| | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, No. 399, Wanyuan Rd., Minhang District, Shanghai, China
| | - Kuiran Dong
- Surgical Department, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Gu
- Nursing Department, Children's Hospital of Fudan University, Shanghai, China
| | - Jie Chen
- Surgical Department/Stoma Wound Care Clinic, Children's Hospital of Fudan University, Shanghai, China
| | - Zhuowen Yu
- Department of Gastroenterology, Children's Hospital of Fudan University, No. 399, Wanyuan Rd., Minhang District, Shanghai, China.
| |
Collapse
|
13
|
Weintraub Y, Collen LV, Hussey S, Mitrova K, Machta JS, Kang B, Granot M, D'Arcangelo G, Spencer EA, Kolho KL, Yeh PJ, Sladek M, Scarallo L, Palomino L, Afzal NA, de Laffolie J, Miele E, Bramuzzo M, Olén O, Russell RK, Rohani P, Tzivinikos C, Urlep D, van Rheenen PF, de Ridder L, Yogev D, Schneider AM, Cohen S, Garcia-Romero R, Dipasquale V, Uhlig HH, Shouval DS. Effectiveness and Safety of Adalimumab in Patients With Very Early-Onset Inflammatory Bowel Disease: A Retrospective Study on Behalf of the Porto Inflammatory Bowel Disease Working Group of European Society for Pediatric Gastroenterology Hepatology and Nutrition. Inflamm Bowel Dis 2025:izae302. [PMID: 39813158 DOI: 10.1093/ibd/izae302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND AND AIMS Patients with very early-onset inflammatory bowel disease (VEO-IBD), with an age of onset < 6 years, can present with severe manifestations and may require biologic therapy. Infliximab and adalimumab are approved for induction and maintenance in pediatric IBD patients but are licensed only above the age of 6 years. Effectiveness and safety data on adalimumab in this patient population are lacking. We assessed the therapeutic response to help close this gap. METHODS This retrospective study involved 30 sites worldwide. Demographic, clinical, and laboratory data were collected from patients with VEO-IBD who commenced adalimumab therapy before the age of 6 years. RESULTS Seventy-eight patients (37 Crohn's disease, 26 ulcerative colitis, and 15 with IBD-unclassified) were included. Median age of IBD onset was 2.6 (1.3-4.1) years, with 30 (38.5%) patients diagnosed at age <2 years. Median age at adalimumab initiation was 4.2 (2.8-5.1) years. Adalimumab was used as second-line biologic therapy in 45 (57.7%) patients after infliximab. The median time to last follow-up was 63 (22-124) weeks. Significant improvement in clinical scores, CRP, fecal calprotectin, and weight Z-score were observed by Week 52. Adalimumab durability rates were 61.9%, 48.1%, and 35.6% after 1, 2, and 3 years, respectively. Drug discontinuation rates were not dependent on IBD type, age, prior anti-TNF exposure, or concomitant immunomodulatory treatment. Four (5.1%) patients developed serious infections, including 1 patient with TTC7A deficiency who died following adenovirus sepsis. CONCLUSION Adalimumab therapy is a viable therapeutic option in patients with VEO-IBD with an acceptable safety profile.
Collapse
Affiliation(s)
- Yael Weintraub
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva 4920235, Israel
- Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lauren V Collen
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Seamus Hussey
- Department of Paediatrics, University of Medicine and Health Sciences, RCSI, Dublin 2, Ireland
- Department of Paediatrics, University College Dublin, Dublin 4, Ireland
| | - Katarina Mitrova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Joseph S Machta
- Paediatric Gastroenterology, Royal London Children's Hospital, Barts Health NHS Trust, London, E1 1FR, UK
| | - Ben Kang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Maya Granot
- Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Pediatric Gastroenterology and Nutrition Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 5262100, Israel
| | - Giulia D'Arcangelo
- Department of Women's and Children's Health Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome-Umberto I Hospital, 00185 Rome, Italy
| | - Elizabeth A Spencer
- Icahn School of Medicine, Mount Sinai, Division of Pediatric Gastroenterology & Nutrition, Department of Pediatrics, New York, NY 10029, USA
| | - Kaija-Leena Kolho
- Department of Pediatric Gastroenterology, Children's Hospital, University of Helsinki and HUS, 00290 Helsinki, Finland
| | - Pai-Jui Yeh
- Translational Gastroenterology Unit, University of Oxford, Oxford, OX1 2JD, UK
| | - Malgorzata Sladek
- Department of Pediatrics, Gastroenterology and Nutrition, University Children's Hospital, Jagiellonian University Medical College, 31-008 Cracow, Poland
| | - Luca Scarallo
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
- Department of NEUROFARBA, University of Florence, 50121 Florence, Italy
| | - Laura Palomino
- Gastroenterology and Nutrition Department, Hospital Infantil Universitario Niño Jesús, 28009 Madrid, Spain
| | - Nadeem Ahmad Afzal
- Department of Paediatric Gastroenterology, Southampton Children's Hospital, Southampton, SO16 6YD, UK
| | - Jan de Laffolie
- General Pediatrics & Pediatric Gastroenterology, Justus Liebig University, 35390 Giessen, Germany
| | - Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," 80131 Naples, Italy
| | - Matteo Bramuzzo
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo," 34137 Trieste, Italy
| | - Ola Olén
- Division of Clinical Epidemiology, Department of Medicine Sona, Karolinska Institutet, 171 76 Stockholm, Sweden
- Pediatric Gastroenterology and Nutrition Unit, Sachs' Children's Hospital, 171 77 Stockholm, Sweden
| | - Richard K Russell
- Department of Paediatric Gastroenterology, Royal Hospital for Children and Young People, Edinburgh, EH16 4TJ, UK
| | - Pejman Rohani
- Pediatric Gastroenterology and Hepatology Research Center, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, 14197 33151 Tehran, Islamic Republic of Iran
| | - Christos Tzivinikos
- Paediatric Gastroenterology Department, Al Jalila Children's Specialty Hospital, Mohammed Bin Rashid University, Dubai Medical College, Dubai, United Arab Emirates
| | - Darja Urlep
- Department of Gastroenterology, Hepatology and Nutrition, Children's Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Patrick F van Rheenen
- Department of Paediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Lissy de Ridder
- Department of Paediatric Gastroenterology, Erasmus University Medical Center, Sophia Children's Hospital, 3015 GD Rotterdam, The Netherlands
| | - Dotan Yogev
- Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Anna-Maria Schneider
- Department of Pediatrics, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Shlomi Cohen
- Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Pediatric Gastroenterology Institute, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | - Ruth Garcia-Romero
- Pediatric Gastroenterology and Nutrition Unit, Miguel Servet Hospital, 50009 Zaragoza, Spain
| | - Valeria Dipasquale
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi," University Hospital "G. Martino," 98124 Messina, Italy
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, OX1 2JD, UK
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, OX3 9DU, UK
- Department of Pediatrics, University of Oxford, Oxford, OX3 9DU, UK
| | - Dror S Shouval
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva 4920235, Israel
- Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
14
|
Watson A, Harris RA, Engevik AC, Oezguen N, Nicholson MR, Dooley S, Stubler R, Satter LF, Karam LB, Kellermayer R. MYO5B and the Polygenic Landscape of Very Early-Onset Inflammatory Bowel Disease in an Ethnically Diverse Population. Inflamm Bowel Dis 2025; 31:189-199. [PMID: 39096520 DOI: 10.1093/ibd/izae169] [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: 04/23/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Genetic discovery in very early-onset inflammatory bowel disease (VEO-IBD) can elucidate not only the origins of VEO-IBD, but also later-onset inflammatory bowel disease. We aimed to investigate the polygenic origins of VEO-IBD in a cohort with a high proportion of Hispanic patients. METHODS Patients with VEO-IBD who underwent whole exome sequencing at our center were included. Genes were categorized as genes of interest (GOIs) (129 genes previously described to be associated with VEO-IBD) or non-GOIs. VEO-IBD "susceptibility" single nucleotide variants (SNVs) were identified through enrichment compared with gnomAD (Genome Aggregation Database) and ALFA (Allele Frequency Aggregator) and were scored by Combined Annotation Dependent Depletion for deleteriousness. Gene networks carrying susceptibility SNVs were created. Myosin 5b immunofluorescence was also studied. RESULTS Fifty-six patients met inclusion criteria, and 32.1% identified as Hispanic. Monogenic disease was infrequent (8.9%). Significant enrichment of GOI susceptibility SNVs was observed, notably in MYO5B, especially in Hispanics. MEFV, TNFAIP3, SH3TC2, and NCF2 were also central participants in the GOI networks. Myosin 5b immunofluorescence in colonic mucosa was significantly reduced in those with MYO5B susceptibility SNVs compared with control subjects. Seven genes (ESRRA, HLA-DQ1, RETSAT, PABPC1, PARP4, CCDC102A, and SUSD2) were central participants in the non-GOI networks. CONCLUSIONS Our results support the polygenic nature of VEO-IBD, in which key participants, like MYO5B, were identified through network analytics. Rare variant load within susceptibility genes may be relevant not only for the genetic origins of inflammatory bowel disease, but also for the age of disease onset. Our findings could guide future work in precision medicine.
Collapse
Affiliation(s)
- Ashleigh Watson
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - R Alan Harris
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Amy C Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Numan Oezguen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, USA
| | - Maribeth R Nicholson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sarah Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Rachel Stubler
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Lisa Forbes Satter
- Department of Pediatric Allergy and Immunology, William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Lina B Karam
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Richard Kellermayer
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Children's Nutrition and Research Center, U.S. Department of Agriculture Agricultural Research Service, Houston, TX, USA
| |
Collapse
|
15
|
Jølving LR, Zegers FD, Lund K, Wod M, Nielsen J, Qvist N, Nielsen RG, Nørgård BM. Children and Adolescents Diagnosed With Inflammatory Bowel Disease Are at Increased Risk of Developing Diseases With a Possible Autoimmune Pathogenesis. Inflamm Bowel Dis 2025; 31:87-94. [PMID: 38507606 DOI: 10.1093/ibd/izae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Indexed: 03/22/2024]
Abstract
BACKGROUND The development of diseases with a possible autoimmune pathogenesis is common in adults with inflammatory bowel disease (IBD). In early onset IBD, it may differ but the evidence is sparse. We aimed to investigate the risk and time span from IBD diagnosis to outcomes with different associated disorders with possible autoimmune pathogenesis. METHODS A register-based study included all Danish patients with early onset of IBD (≤18 years) between 1980 and 2021 and 50 matched references without IBD for each case. We examined the risk of type 1 and type 2 diabetes, celiac disease, thyroid disease, rheumatoid arthritis, psoriatic arthritis, and spondyloarthritis in Cox regression models. RESULTS In total, 6822 patients with IBD were identified, and 337 728 matched references. The median age at the time of IBD diagnosis or index date for the matched references was 16 years (25-75 percentile: 13-18 years), and the median age at the time of an outcome or at the end of follow-up was 28.1 years (25-75 percentile: 21.5-37.0 years). According to the cumulative incidence plots psoriatic arthritis, and spondyloarthritis was diagnosed approximately 10 years after the IBD onset, and the remaining outcomes later. The adjusted hazard ratio after full follow-up was 4.72 (95% CI, 3.85-5.80) for psoriatic arthritis, 5.21 (95% CI, 4.17-6.50) for spondyloarthritis, 2.77 (95% CI, 1.92-4.00) for celiac disease, 2.15 (95% CI, 1.54-3.01) for rheumatoid arthritis, 1.69 (95% CI, 1.23-2.32) and 1.64 (95% CI, 1.21-2.21) for type 1 and type 2 diabetes, respectively. For thyroid disease, it was 1.16 (95% CI, 0.97-1.40). CONCLUSIONS The risk estimates were significantly increased for all outcomes at the end of follow-up, except for thyroid disease, but according to the cumulative incidence plots, only psoriatic arthritis and spondyloarthritis occurred earlier in the IBD cohort than in the matched references.
Collapse
Affiliation(s)
- Line Riis Jølving
- Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Epidemiology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Ken Lund
- Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Epidemiology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mette Wod
- Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Epidemiology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jan Nielsen
- Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Epidemiology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Niels Qvist
- Research Unit for Surgery and Center for IBD Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Centre of Excellence in Gastrointestinal Diseases and Malformations in Infancy and Childhood (GAIN), Odense University Hospital, Odense, Denmark
| | - Rasmus Gaardskær Nielsen
- Centre of Excellence in Gastrointestinal Diseases and Malformations in Infancy and Childhood (GAIN), Odense University Hospital, Odense, Denmark
- Hans Christian Andersen Children's Hospital, Odense University Hospital, and Research Unit of Pediatrics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bente Mertz Nørgård
- Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Epidemiology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
16
|
Kaplina AV, Petrova NA, Pervunina TM, Khavkin AI, Surkov AN, Nazarenko LP, Getmanov SD, Sitkin SI. Necrotizing Enterocolitis: Pathogenetic Features and Differential Diagnosis with Inflammatory Bowel Disease in Newborns. CURRENT PEDIATRICS 2025; 23:438-446. [DOI: 10.15690/vsp.v23i6.2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2025]
Abstract
Necrotizing enterocolitis (NEC) is a disease primarily affecting premature infants. NEC pathogenesis is based on the development of inflammation damaging mucous membranes associated with bacterial colonization, intestinal epithelium immaturity, intestinal blood flow regulation, and excessive inflammatory response activation. Inflammatory bowel disease (IBD) with very early onset (VEO-IBD) can also manifest in the neonatal period. They are characterized by severe course, often resistant to traditional immunosuppressive therapy. This article discusses the features of NEC pathogenesis and differential diagnosis with VEO-IBD. Despite certain similarities in pathogenesis, NEC and IBD are different diseases. Infantile onset IBD is more often associated with monogenic diseases and primary immunodeficiency. VEO-IBD is a chronic disease characterized by damage to all intestinal layers and has a lower incidence compared to NEC. Its clinical manifestations may include chronic diarrhea, blood in stool, delayed physical development, perianal diseases, and ulcerations in the oral cavity. Infantile onset VEO-IBD usually affects the colon, while NEC affects the ileum in premature infants. The intestinal microbiome in VEO-IBD also has specific features. It has been reported that clinical cases of Crohn’s disease in patients who had NEC in the neonatal period are associated with NEC surgery. It is crucial to consider perinatal period features when assessing the IBD risk (prenatal effects of antibacterial therapy and smoking, several courses of antibacterial therapy during the first year of life, and formula feeding).
Collapse
Affiliation(s)
| | | | | | - Anatoly I. Khavkin
- Research Clinical Institute of Childhood; Belgorod National Research University
| | - Andrey N. Surkov
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University
| | | | | | - Stanislav I. Sitkin
- Almazov National Medical Research Centre; North-Western State Medical University named after I.I. Mechnikov; Institute of Experimental Medicine
| |
Collapse
|
17
|
Schwerd T. [Inflammatory bowel diseases in children and adolescents : An overview with particular attention to genetic testing]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:31-39. [PMID: 39774688 DOI: 10.1007/s00108-024-01827-8] [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: 11/27/2024] [Indexed: 01/11/2025]
Abstract
Pediatric-onset inflammatory bowel disease (PIBD) is increasingly recognized in Germany. Patients with PIBD often present with more extensive and active disease. Clinical suspicion of IBD requires early initiation of the diagnostic work-up (e.g., non-invasive fecal marker for inflammation) and referral to a pediatric gastroenterology center. In the presence of very early-onset IBD, as well as further criteria such as family history, relevant comorbidities, and extraintestinal manifestations, genetic testing for monogenic forms of IBD should be considered. The aim of treatment is to normalize quality of life and prevent bowel damage and complications, thereby enabling normal physical, social, and emotional development of the child. The selection of treatment is based on individual risk stratification, which considers disease severity and activity. PIBD patients often receive more intensified therapies, including biologics and small molecules. However, anti-tumor necrosis factor (TNF) antibodies are the only approved biologics for PIBD (above the age of 6 years). Therefore, licensed anti-TNF is a mainstay of PIBD therapy. Regular PIBD and drug monitoring should be performed according to the treat-to-target approach. Patients with PIBD and their families have special health care needs and require an interdisciplinary team of specialized medical doctors, psychologists, social workers, dieticians, and nurses. Close cooperation between the local pediatrician/family doctor and the pediatric gastroenterologist is important to achieve the long-term goals. Psychosocial consequences are important but are often underestimated.
Collapse
Affiliation(s)
- Tobias Schwerd
- Dr. von Haunersches Kinderspital, Kinderklinik und Kinderpoliklinik, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, München, Deutschland.
| |
Collapse
|
18
|
Abu Shtaya A, Orenstein N, Bazak L, Lidzbarsky G, Kalis ML, Amarilyo G, Sofrin-Drucker E, Jaron R, Shahar NR, Gilad NK, Basel-Salmon L. High frequency of MEFV disease-causing variants in children with very-early-onset inflammatory bowel disease. Pediatr Res 2025; 97:268-272. [PMID: 38734812 DOI: 10.1038/s41390-024-03242-z] [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] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 03/15/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Biological similarities between inflammatory bowel disease (IBD) and familial Mediterranean fever (FMF) have been described in humans and animal models suggesting a possible common genetic basis. FMF is caused by variants in the MEFV gene which encodes pyrin, an immune regulator. This study aimed to investigate the carrier rate of disease-causing MEFV variants in children of different ethnicities diagnosed with very-early-onset IBD (VEO-IBD). METHODS The study included 23 children diagnosed with VEO-IBD who had undergone whole exome sequencing. The exomes were evaluated for MEFV monoallelic and biallelic disease-causing variants and compared to exome sequencing data of 250 probands with suspected monogenic diseases other than IBD. RESULTS Of the 23 children diagnosed with VEO-IBD, 12 (52%) were carriers of at least one MEFV disease-causing variant, which was threefold higher than in individuals without IBD. The most frequent variants identified were p.M694V and p.E148Q (42% each). The allelic frequency of MEFV variants was found to be higher across the VEO-IBD group in 13 of 14 ethnicities compared to the control group. CONCLUSION The study suggests that disease-causing variants in the MEFV gene should be sought in cases of VEO-IBD. However, the clinical importance of this finding is yet to be defined. IMPACT There are biological similarities between inflammatory bowel disease and familial Mediterranean fever, suggesting a possible genetic relationship. Children less than 6 years old clinically diagnosed with inflammatory bowel disease have a threefold higher rate of disease-causing variants in the MEFV gene than controls. Monogenic testing in children with very-early-onset inflammatory bowel disease should include a search for MEFV variants.
Collapse
Affiliation(s)
- Aasem Abu Shtaya
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel.
- Unit of Gastroenterology, Carmel Medical Center, Haifa, Israel.
| | - Naama Orenstein
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lily Bazak
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel
| | - Gabriel Lidzbarsky
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel
| | - Marina Lifshitc Kalis
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel
| | - Gil Amarilyo
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Rheumatology Department, Schneider Children's Medical Centre, Petach Tikvah, Israel
| | - Efrat Sofrin-Drucker
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ranit Jaron
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Noa Ruhrman Shahar
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel
| | - Nesia Kropach Gilad
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Basel-Salmon
- Recanati Genetic Institute, Rabin Medical Center-Beilinson Hospital, Petach Tikva, Israel
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
19
|
Xu X, Gao Y, Xiao Y, Yu Y, Huang J, Su W, Li N, Xu C, Gao S, Wang X. Characteristics of the gut microbiota and the effect of Bifidobacterium in very early-onset inflammatory bowel disease patients with IL10RA mutations. Front Microbiol 2024; 15:1479779. [PMID: 39687875 PMCID: PMC11647010 DOI: 10.3389/fmicb.2024.1479779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Very early-onset inflammatory bowel disease (VEO-IBD) is a distinct subtype of inflammatory bowel disease (IBD) characterized by onset before the age of 6 years, and patients often exhibit more severe clinical features. Interleukin 10 receptor alpha (IL10RA) is a hotspot mutation in the Chinese population and is associated with a poor prognosis closely linked to the onset of IBD. However, limited knowledge exists regarding how the IL10RA mutation influences the host microbiota and its role in disease development. We employed 16S rRNA sequencing to conduct a comprehensive assessment of microbial changes in different types of IBD, employed database to thoroughly examine the influence of Bifidobacterium in IBD and to demonstrate a potential positive effect exerted by Bifidobacterium breve M16V (M16V) through a mouse model. The study demonstrated a significant reduction in the abundance and diversity of the gut microbiota among children with IL10RA mutations compared to those with late-onset pediatric IBD and nonmutated VEO-IBD. Furthermore, the analysis identified genera capable of distinguishing between various types of IBD, with the genus Bifidobacterium emerging as a potential standalone diagnostic indicator and Bifidobacterium may also be involved in related pathways that influence the progression of IBD, such as the biosynthesis of amino acids and inflammation-related pathways. This study corroborated the efficacy of Bifidobacterium in alleviating intestinal inflammation. The impact of IL10RA mutations on VEO-IBD may be mediated by alterations in microbes. M16V demonstrates efficacy in alleviating colitis and holds promise as a novel microbial therapy.
Collapse
Affiliation(s)
- Xu Xu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanqi Gao
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Xiao
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiebin Huang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Su
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Li
- Department of Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chundi Xu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenshen Gao
- Clinical Research and Development Center of Shanghai Municipal Hospitals, Shanghai Shenkang Hospital Development Center, Shanghai, China
| | - Xinqiong Wang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Grotra R, Karri PS, Gupta A, Malik R, Gupta AK, Meena JP, Seth R. Matched Unrelated Donor Hematopoietic Stem Cell Transplant as Successful Curative Therapy for IL10RB Mutation-Associated Very Early Onset IBD. Pediatr Transplant 2024; 28:e14891. [PMID: 39539152 DOI: 10.1111/petr.14891] [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: 07/09/2024] [Revised: 10/17/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Inflammatory bowel diseases are complex chronic disorders with a relapsing-remitting course that affect the gut due to dysregulated immune response. The incidence of these disorders is increasing globally along with an increase in the incidence in pediatric population. Very early onset inflammatory bowel diseases are seen in children with age less than 6 years, where monogenic causes predominate. With the advent of next-generation sequencing methods, these disorders are being diagnosed more. Interleukin-10 receptor mutation-associated inflammatory bowel diseases is one such monogenic disorder where immunosuppression shows poor response. METHODS We report the case of an 8-month-old child of Indian origin who presented with severe enterocolitis and rectovaginal fistulas. She was evaluated on lines of a very early onset inflammatory bowel disease. She was found to have a mutation in the interleukin-10 receptor causing severe enterocolitis. She underwent a diversion colostomy. She was admitted at 25 months of age for the hematopoietic-stem-cell-transplant (HSCT). The conditioning regimen used consisted of busulfan, fludarabine, and anti-thymocyte-globulin (ATG). The child received a 10/10 human leukocyte antigen (HLA) matched from a matched-unrelated adult female donor with bone marrow stem cell product at a dose of 5.6 million CD34+ cells per kg. RESULTS She was treated successfully by a matched unrelated donor HSCT. At present, she is 2 years and 4 months posttransplant and is cured. CONCLUSIONS Early recognition and prompt genetic testing can help in diagnosing and establishing the cause of a very early onset inflammatory bowel disease. Very early onset inflammatory bowel disease caused due to interleukin-10 receptor mutations can be cured by HSCT.
Collapse
Affiliation(s)
- Rohan Grotra
- Division of Pediatric Gastroenterology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Padma Sagarika Karri
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Aditya Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rohan Malik
- Division of Pediatric Gastroenterology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rachna Seth
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
21
|
Bramuzzo M, Cananzi M, Alvisi P, Cardile S, Romano C, Aloi M, Arrigo S, Felici E, Lonoce L, Pieri ES, Scarallo L, Strisciuglio C, Di Siena A, Lega S. Hemophagocytic Lymphohistiocytosis/Macrophage Activation Syndrome in pediatric Inflammatory Bowel Disease: clinical characteristics and outcomes. Eur J Pediatr 2024; 183:5411-5418. [PMID: 39404873 DOI: 10.1007/s00431-024-05772-4] [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: 06/19/2024] [Revised: 09/01/2024] [Accepted: 09/07/2024] [Indexed: 11/01/2024]
Abstract
Hemophagocytic Lymphohistiocytosis (HLH)/Macrophage Activation Syndrome (MAS) in children with inflammatory bowel disease (IBD) has been reported only anecdotally. This study aimed at describing the clinical features and outcomes of children diagnosed with both IBD and HLH/MAS. Data on IBD and HLH/MAS characteristics, biochemical, microbiological and genetic assessments, treatments, and outcomes were collected from the Italian Pediatric IBD Registry and presented using descriptive statistics. Out of 4643 patients with IBD, 18 (0.4%) were diagnosed with HLH/MAS, including 12 with ulcerative colitis and 6 with Crohn disease. Among the 18 patients, 7 (39%) had early-onset IBD, but the median age at HLH/MAS diagnosis was 14.0 years (IQR 11.9-16.0). Half of the patients had active IBD at HLH/MAS diagnosis, 11 (61%) patients were on thiopurines, and 6 (33%) were on anti-TNF biologics. An infectious trigger was identified in 15 (83%) patients. One (5%) patients was diagnosed with XIAP deficiency. All patients discontinued thiopurines and 5 (83.3%) discontinued anti-TNF biologics; 16 (80%) patients received steroids for HLH/MAS. Three (17%) patients had a relapse of HLH/MAS. No patient developed lymphoma or died during a median follow-up of 2.7 years (IQR 0.8-4.4). Conclusions: HLH/MAS mainly affects children with early-onset IBD but primarily develops during adolescence, following an infection while on immunosuppressant treatment. Although the prognosis is generally favorable, it is crucial to investigate an underlying immune deficiency.
Collapse
Affiliation(s)
- Matteo Bramuzzo
- Institute for Maternal and Child Health (IRCCS) "Burlo Garofolo", Via Dell' Istria 65, 34137, Trieste, Italy.
| | - Mara Cananzi
- Unit of Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child With Liver Transplantation, Dpt. of Women's and Children's Health, University Hospital of Padova, Padua, Italy
| | - Patrizia Alvisi
- Pediatric Gastroenterology Unit, Maggiore "CA Pizzardi" Hospital, Bologna, Italy
| | - Sabrina Cardile
- Gastroenterology, Digestive Endoscopy and Nutrition Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina, Italy
| | - Marina Aloi
- Pediatric Gastroenterology and Hepatology Unit, Sapienza University of Rome, Umberto I Hospital, Rome, Italy
| | - Serena Arrigo
- Pediatric Gastroenterology and Endoscopy, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Felici
- Pediatric and Pediatric Emergency Unit, Children Hospital, AOU SS Antonio E Biagio E C. Arrigo, Alessandria, Italy
| | - Luisa Lonoce
- Clinical Pediatrics, Department of Molecular Medicine and Development, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
| | | | - Luca Scarallo
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Di Siena
- Division of Pediatrics, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Sara Lega
- Institute for Maternal and Child Health (IRCCS) "Burlo Garofolo", Via Dell' Istria 65, 34137, Trieste, Italy
| |
Collapse
|
22
|
Alcocer Alkureishi L, Hageman JR, Biank V. Diagnosing and Managing Inflammatory Bowel Disease in Young Children. Pediatr Ann 2024; 53:e433-e434. [PMID: 39653341 DOI: 10.3928/19382359-20241022-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Shankar S, Majumder S, Mukherjee S, Bhaduri A, Kasturi R, Ghosh S, Iacucci M, Shivaji UN. Inflammatory bowel disease: a narrative review of disease evolution in South Asia and India over the last decade. Therap Adv Gastroenterol 2024; 17:17562848241258360. [PMID: 39575157 PMCID: PMC11580062 DOI: 10.1177/17562848241258360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 11/24/2024] Open
Abstract
The rapid emergence of inflammatory bowel disease (IBD) in Asia in the last two decades is anticipated to pose significant challenges to the healthcare systems of developing countries including India. Several epidemiological factors in the Asia Pacific region have been explored as risk factors for the development of IBD. In this narrative review, we discuss the evolution of adult-onset and paediatric IBD in South Asia and India, in relation to the current global epidemiology, over the last decade. The focus lies on the changing epidemiological landscape of IBD in Asia which signals a paradigm shift in the disease trajectory of a chronic, relapsing, complex disease. We enumerate the disease burden of IBD in India and Asia, analyse the risk factors for its recent rise in incidence and briefly discuss the unique entity of very early-onset IBD. We also list the locoregional challenges in diagnosis and management along with suggestions to overcome them. We highlight the lacunae in data which warrants further research. The anticipated infrastructural challenges and disease evolution are likely to be similar in most newly industrialized countries across South Asia. A combined effort led by IBD experts in the region to understand the true disease burden is important. A strong collaborative network on research and formulation of preventive strategies relevant to the region will help reduce the burden in the future.
Collapse
Affiliation(s)
- Sahana Shankar
- Division of Paediatric Gastroenterology, Department of Paediatrics, Mazumdar Shaw Medical Center, NH Health City, Bangalore, India
| | - Snehali Majumder
- Department of Clinical Research, Narayana Hrudayalaya, NH Health City, Bangalore, India APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Suparna Mukherjee
- Department of Clinical Nutrition and Dietetics, Narayana Hrudayalaya, NH Health City, Bangalore, India
| | | | - Rangarajan Kasturi
- Department of Gastroenterology, Mazumdar Shaw Medical Center, a Unit of Narayana Health, Bangalore, India
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Uday N. Shivaji
- Institute of Immunology and Immunotherapy, University of Birmingham, 2nd Floor, Institute of Translational Medicine, Heritage Building, Mindelsohn Way, Birmingham B15 2TH, UK Department of Gastroenterology, Mazumdar Shaw Medical Center, a Unit of Narayana Health, Bangalore, India
| |
Collapse
|
24
|
You Y, Tao Y, Xu Y, Cao Y, Feng H, Wu Q, Wang Y, Yan W. Clinical analysis and identification of pediatric patients with colonic ulceration. BMC Pediatr 2024; 24:697. [PMID: 39487408 PMCID: PMC11529313 DOI: 10.1186/s12887-024-05174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND A wide variety of diseases mimic inflammatory bowel disease (IBD). This study aimed to reduce the misdiagnosis among children with colonic ulcers. METHODS Eighty-six pediatric patients with colonic ulcers detected by colonoscopy were enrolled in the retrospective study. Children were divided into different groups according to the final diagnosis. The clinical characteristics, laboratory examinations, endoscopic findings, and histopathological results were compared. RESULTS IBD (n = 37) was just responsible for 43% of patients with colonic ulceration. Other diagnosis included autoimmune diseases (n = 9), infectious enteritis (n = 13), gastrointestinal allergy (n = 8), and other diseases (n = 19). Comparing IBD and non-IBD groups, children with IBD had a higher frequency of symptoms like weight loss/failure to thrive (P < 0.001), perianal lesions (P = 0.001), and oral ulcers (P = 0.022), and higher expression levels of platelet (P = 0.006), neutrophil-to-lymphocyte ratio (NLR) (P = 0.001), erythrocyte sedimentation rate (P < 0.001), C-reactive protein (P < 0.001), Immunoglobulin G (P = 0.012), Interleukin-1β (P = 0.003), Interleukin-6 (P = 0.024) and TNF-α (P = 0.026), and a wider ulcer distribution in the lower gastrointestinal tract (LGIT) (P < 0.001). Expression levels of hemoglobin (P < 0.001) and albumin (P = 0.001) were lower in IBD patients. Multivariate analysis showed hemoglobin, NLR, Score of ulceration in LGIT, and pseudopolyps contributing to the diagnosis of pediatric IBD with colonic ulcers. CONCLUSIONS We displayed potential indicators to help diagnose pediatric IBD differentiating from other disorders with colonic ulcers more prudently.
Collapse
Affiliation(s)
- Yaying You
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China
| | - Yijing Tao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China
| | - Yanwen Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Cao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China
| | - Haixia Feng
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China
| | - Qingqing Wu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| | - Weihui Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
25
|
Vuijk SA, Camman AE, de Ridder L. Considerations in Paediatric and Adolescent Inflammatory Bowel Disease. J Crohns Colitis 2024; 18:ii31-ii45. [PMID: 39475081 PMCID: PMC11523044 DOI: 10.1093/ecco-jcc/jjae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 11/02/2024]
Abstract
The incidence of inflammatory bowel disease [IBD] is rising most rapidly among children and adolescents. Paediatric-onset IBD is associated with a more extensive and severe disease course compared to adult-onset IBD. At a young age, screening for underlying genetic and immunological disorders is important and may impact treatment management. Early and effective treatment is crucial to reach disease remission and prevent complications of ongoing active disease. In children with Crohn's disease, exclusive enteral nutrition is an effective induction therapy. Other promising dietary therapies, such as the Crohn's disease exclusion diet, are emerging. Within paediatric IBD, anti-tumour necrosis factor therapy is the only approved biological thus far and additional treatment options are crucially needed. Other biological therapies, such as vedolizumab and ustekinumab, are currently prescribed off-label in this population. A specific challenge in paediatric IBD is the unacceptable and major delay in approval of drugs for children with IBD. A guided transfer period of paediatric patients to adult care is associated with improved disease outcomes and is required. Major knowledge gaps and challenges within paediatric IBD include the aetiology, diagnostics, and monitoring of disease, tailoring of treatment, and both understanding and coping with the physical and psychological consequences of living with IBD. Challenges and research gaps in paediatrics should be addressed without any delay in comparison with the adult field, in order to ensure a high quality of care for all patients with IBD, irrespective of the age of onset.
Collapse
Affiliation(s)
- Stephanie A Vuijk
- Department of Paediatric Gastroenterology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Anouk E Camman
- Department of Paediatric Gastroenterology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Lissy de Ridder
- Department of Paediatric Gastroenterology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Hutmacher F, Doerig S, Grobholz R, Köhler H, Meyer P, Baumann P. Aphthous lesions turned out to be neonatal very early-onset inflammatory bowel disease: a case report. Front Pediatr 2024; 12:1433852. [PMID: 39539768 PMCID: PMC11557482 DOI: 10.3389/fped.2024.1433852] [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: 05/16/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Neonatal diagnosis of inflammatory bowel disease (IBD) proves challenging due to its non-specific symptoms. A term-born neonate showing states of inflammation and aphthae was treated for sepsis and candidiasis before being diagnosed with interleukin-10 receptor deficiency and consecutive IBD. The patient was finally successfully treated by stem cell transplantation. The case illustrates the difficulties of the diagnostic course in IBD as it may mimic other diseases and emphasizes the importance of considering rare differential diagnoses early in the diagnostic process.
Collapse
Affiliation(s)
- Felix Hutmacher
- Department of Neonatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Selina Doerig
- Department of Neonatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Rainer Grobholz
- Medical Faculty, University of Zurich, Zurich, Switzerland
- Institute of Pathology, Kantonsspital Aarau, Aarau, Switzerland
| | - Henrik Köhler
- Medical Faculty, University of Basel, Basel, Switzerland
- Department of Gastroenterology, Kantonsspital Aarau, Aarau, Switzerland
| | - Philipp Meyer
- Department of Neonatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Philipp Baumann
- Department of Neonatology, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
27
|
Zhang H, Zhang M, Zheng X, Xu X, Zheng J, Hu Y, Mei Y, Liu Y, Liang Y. Solid-State Fermentation of Wheat Bran with Clostridium butyricum: Impact on Microstructure, Nutrient Release, Antioxidant Capacity, and Alleviation of Ulcerative Colitis in Mice. Antioxidants (Basel) 2024; 13:1259. [PMID: 39456512 PMCID: PMC11504992 DOI: 10.3390/antiox13101259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigated the effects of solid-state fermentation with Clostridium butyricum on the microstructure of wheat bran, the release of dietary fiber and phenolic compounds, and antioxidant capacity. Compared with unfermented wheat bran, insoluble dietary fiber and phytic acid content decreased, whereas soluble dietary fiber and water-extractable arabinoxylan content increased in C. butyricum culture. Because of the increased release of phenolic compounds, such as ferulic acid and apigenin, and organic acids, such as isobutyric acid, the antioxidant capacity of the culture was considerably improved. Furthermore, the culture of C. butyricum treated with dextran sulfate sodium-induced ulcerative colitis in mice enhanced the expression of intestinal mucus and tight-junction proteins, modulating the gut microbiota structure, increasing the levels of short-chain fatty acids in the intestine, and restoring the essential functions of the gut microbiota. These anti-inflammatory effects stemmed from the combined action of various effective components.
Collapse
Affiliation(s)
- Heng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Min Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Xin Zheng
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Xiaofang Xu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Jiawen Zheng
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Yuanliang Hu
- Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, College of Life Sciences, Hubei Normal University, Huangshi 435002, China;
| | - Yuxia Mei
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| | - Yangyang Liu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Yunxiang Liang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.Z.); (M.Z.); (X.Z.); (X.X.); (J.Z.); (Y.M.)
| |
Collapse
|
28
|
Catassi G, D'Arcangelo G, Norsa L, Bramuzzo M, Hojsak I, Kolho KL, Romano C, Gasparetto M, Di Giorgio A, Hussey S, Yerushalmy-Feler A, Turner D, Matar M, Weiss B, Karoliny A, Alvisi P, Tzivinikos C, Aloi M. Outcome of Very Early Onset Inflammatory Bowel Disease Associated With Primary Sclerosing Cholangitis: A Multicenter Study From the Pediatric IBD Porto Group of ESPGHAN. Inflamm Bowel Dis 2024; 30:1662-1669. [PMID: 37768032 DOI: 10.1093/ibd/izad218] [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: 07/20/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Whether primary sclerosing cholangitis related to inflammatory bowel disease (PSC-IBD) diagnosed before 6 years (ie, VEO-IBD) has a distinct phenotype and disease course is uninvestigated. We aimed to analyze the characteristics and natural history of VEO-PSC-IBD, compared with early and adolescent-onset PSC-IBD. METHODS This is a multicenter, retrospective, case-control study from 15 centers affiliated with the Porto and Interest IBD group of ESPGHAN. Demographic, clinical, laboratory, endoscopic, and imaging data were collected at baseline and every 6 months. Inflammatory bowel disease-related (clinical remission, need for systemic steroids and biologics, and surgery) and PSC-related (biliary and portal hypertensive complications, need for treatment escalation and liver transplantation, cholangiocarcinoma, or death) outcomes were compared between the 2 groups. RESULTS Sixty-nine children were included, with a median follow-up of 3.63 years (interquartile range, 1-11): 28 with VEO-PSC-IBD (23 UC [82%], 2 IBD-U [7%] and 3 [11%] CD), and 41 with PSC-IBD (37 UC [90%], 3 IBDU [7.5%] and 1 [2.5%] CD). Most patients with UC presented with pancolitis (92% in VEO-PSC-UC vs 85% in PSC-UC, P = .2). A higher number of patients with VEO-PSC-IBD were diagnosed with PSC/autoimmune hepatitis overlap syndrome than older children (24 [92%] vs 27 [67.5%] PSC-IBD, P = .03), whereas no other differences were found for PSC-related variables. Time to biliary strictures and infective cholangitis was lower in the VEO-PSC-IBD group (P = .01 and P = .04, respectively), while no difference was found for other outcomes. No cases of cholangiocarcinoma were reported. CONCLUSIONS Primary sclerosing cholangitis related to inflammatory bowel disease has similar baseline characteristics whether diagnosed as VEO-IBD or thereafter. A milder disease course in terms of biliary complications characterizes VEO-PSC-IBD.
Collapse
Affiliation(s)
- Giulia Catassi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, Rome, Italy
| | - Giulia D'Arcangelo
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, Rome, Italy
| | - Lorenzo Norsa
- Pediatric Hepatology Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Matteo Bramuzzo
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo," Trieste, Italy
| | - Iva Hojsak
- University Children's Hospital Zagreb, University of Zagreb Medical School, Zagreb, Croatia
| | - Kaija-Leena Kolho
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, University of Messina, Messina, Italy
| | - Marco Gasparetto
- Department of Paediatric Gastroenterology, Barts Health Trust, The Royal London Children's Hospital, London, UK
| | - Angelo Di Giorgio
- Pediatric Hepatology Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Seamus Hussey
- National Children's Research Centre, Royal College of Surgeons of Ireland and University College Dublin, Dublin, Ireland
| | - Anat Yerushalmy-Feler
- Pediatric Gastroenterology Institute "Dana-Dwek" Children's Hospital, Tel Aviv University, Tel Aviv, Israel
| | - Dan Turner
- Shaare Zedek Medical Center, the Hebrew University of Jerusalem, Israel
| | - Manar Matar
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Petach Tikva, Israel
| | - Batia Weiss
- Pediatric Gastroenterology and Cystic Fibrosis Unit, University of Messina, Messina, Italy
| | - Anna Karoliny
- Heim Pal National Pediatric Institute, Budapest, Hungary
| | - Patrizia Alvisi
- Pediatric Unit, Maggiore Hospital, Largo Bartolo Nigrisoli, 2, 40133 Bologna, Italy
| | - Christos Tzivinikos
- Department of Pediatric Gastroenterology, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, Rome, Italy
| |
Collapse
|
29
|
邢 娇, 徐 娟, 刘 志, 程 卫. [A case report of Crohn's disease in a child with trisomy 8]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:982-985. [PMID: 39267515 PMCID: PMC11404466 DOI: 10.7499/j.issn.1008-8830.2405069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/08/2024] [Indexed: 09/17/2024]
Abstract
The patient is a 12-year-old male who has experienced recurrent perianal abscesses for over 10 years, along with recurrent oral ulcers and deformities in the joints of hands and feet. Gastrointestinal endoscopy and capsule endoscopy revealed multiple ulcers in the digestive tract. Combined with his histopathological examinations, the patient was diagnosed with Crohn's disease. Whole exome sequencing and peripheral blood karyotype analysis indicated a karyotype of 47,XY,+8. The patient was treated with a "step-up" strategy. His clinical symptoms were under control, with significant improvement observed during endoscopic examination. This case suggests that early-onset inflammatory bowel disease may have genetic susceptibility, and when accompanied by other multi-system involvement, the possibility of chromosomal abnormalities, such as trisomy 8, should be considered and given due attention.
Collapse
|
30
|
Collen LV, Mitsialis V, Kim DY, Bresnahan M, Yang J, Tuthill M, Combs A, Barends J, Field M, Liu E, Bearup R, Okoroafor I, Klein C, Muise AM, Bousvaros A, Ouahed J, Snapper SB. Efficacy and Safety of Anti-Tumor Necrosis Factor Alpha in Very Early Onset Inflammatory Bowel Disease. Inflamm Bowel Dis 2024; 30:1443-1453. [PMID: 37847820 PMCID: PMC11369069 DOI: 10.1093/ibd/izad196] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Very early onset inflammatory bowel disease (VEOIBD) is defined as disease onset in patients younger than 6 years. Challenges in treatment of VEOIBD include lack of approved therapies and increased incidence of monogenic immunodeficiencies. We report on patterns of anti-TNF use, efficacy, and safety in a large cohort of patients with VEOIBD. METHODS Very early onset inflammatory bowel disease patients receiving care at a single center were prospectively enrolled in a data registry and biorepository starting in 2012. Whole exome sequencing was available to all patients. Clinical data including IBD medication use and response were extracted from the medical record. We examined antitumor necrosis factor (anti-TNF) cumulative exposure and time to failure and evaluated the effect of covariates on anti-TNF failure using Cox proportional hazard regression. RESULTS In this cohort of 216 VEOIBD patients with median 5.8-year follow-up, 116 (53.7%) were TNF-exposed. Sixty-two TNF-exposed patients (53.4%) received their first dose at younger than 6 years. Cumulative exposure to anti-TNF was 23.6% at 1 year, 38.4% at 3 years, and 43.4% at 5 years after diagnosis. Cumulative exposure was greater in patients with Crohn's disease (P = .0004) and in those diagnosed in 2012 or later (P < .0001). Tumor necrosis factor failure occurred in 50.9% of those exposed. Features predictive of anti-TNF failure included ulcerative colitis/IBD-unclassified (hazard ratio, 1.94; P = .03), stricturing (hazard ratio, 2.20; P = .04), and younger age at diagnosis (hazard ratio, 1.25; P = .01). Adverse events occurred in 22.6% of infliximab-exposed and 14.3% of adalimumab-exposed. CONCLUSIONS Efficacy and safety of anti-TNFs in VEOIBD is comparable to what has previously been reported in older patients.
Collapse
Affiliation(s)
- Lauren V Collen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Vanessa Mitsialis
- Division of Gastroenterology, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - David Y Kim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Mairead Bresnahan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jessica Yang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Margaret Tuthill
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Abigail Combs
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jared Barends
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Field
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Enju Liu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Richelle Bearup
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ibeawuchi Okoroafor
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Klinikum, and Gene Center, Ludwig Maximilians Universität München, Germany
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Athos Bousvaros
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jodie Ouahed
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
31
|
Köglmeier J, Lindley KJ. Congenital Diarrhoeas and Enteropathies. Nutrients 2024; 16:2971. [PMID: 39275287 PMCID: PMC11397474 DOI: 10.3390/nu16172971] [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: 07/25/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Congenital diarrhoeas and enteropathies (CODE) are a heterogeneous group of disorders. Many affected infants present with catastrophic dehydration in the first few days of life, although the clinical phenotype is variable. Advances in the understanding of underlying pathomechanisms and genetic testing, as well as improved management, in particular intravenous nutrition support, have allowed affected patients to survive well beyond childhood. Awareness and understanding of these rare diseases are hence needed, both amongst paediatricians and adult physicians. In this review, we discuss the different groups of disorders based on a review of the current literature and provide a diagnostic and therapeutic approach. Many of the subtypes of CODE result in the need for prolonged or indefinite parenteral nutrition. Further research is needed to identify new CODE to improve the recognition and management of these children, which can assist in developing new targeted therapies and potentially a long-term cure.
Collapse
Affiliation(s)
- Jutta Köglmeier
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
| | - Keith James Lindley
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
| |
Collapse
|
32
|
Santoro L, Grillo F, D'Armiento M, Buccoliero AM, Rocco M, Ferro J, Vanoli A, Cafferata B, Macciomei MC, Mescoli C, Cananzi M, Alaggio R, Fassan M, Mastracci L, Francalanci P, Parente P. Clinicopathologic Features of Primary Immunodeficiency Monogenic Disease-related Very Early Onset Inflammatory Bowel Disease: Focus on Gastrointestinal Histologic Features in IFIH1 Mutations. Adv Anat Pathol 2024:00125480-990000000-00113. [PMID: 39140676 DOI: 10.1097/pap.0000000000000457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Very early onset inflammatory bowel disease (VEO-IBD) is a clinical term referring to IBD-like symptomatology arising in children younger than 6 years. VEO-IBD may be due to polygenic etiology in "pure" IBD (Crohn disease-CD and ulcerative colitis-UC), or it may be caused by primary immunodeficiency underlined by monogenic disease. Primary immunodeficiency monogenic diseases have a Mendelian inheritance and affect the immune system with multiorgan morbidity and possible effects on the gastrointestinal system. Primary Immunodeficiency monogenic diseases differ from "pure" IBD as the latter primarily affect the gastrointestinal tract with mitigated extraintestinal symptomatology. Since their first description, primary immunodeficiency monogenic diseases, although rare, have been the subject of increasing interest due to their dramatic phenotype, difficulty in reaching a timely diagnosis, and specific therapeutic approach. In this paper, we present a brief review of primary immunodeficiency monogenic diseases, focusing on to their clinicopathologic features as well as delving, in greater detail, into monogenic diseases caused by IFIH1 mutations. The clinicopathologic features of 4 patients with IFIH1, a gene involved in interferon pathway deficiency, will be described using a histologic pattern of damage approach confirming the need to avoid the histologic diagnosis of VEO-IBD in children younger than 6 years.
Collapse
Affiliation(s)
- Luisa Santoro
- Pathology Unit, Azienda Ospedaliera Padova, Via Ospedale Vecchio
| | - Federica Grillo
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
| | - Maria D'Armiento
- Pathology Unit, Department of Public Health, University of Naples Federico II, Via Sergio Pansini, Napoli
| | | | - Michele Rocco
- Department of Pathology, Santobono-Pausilipon, Children's Hospital, Naples
| | - Jacopo Ferro
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo, Genova
| | - Alessandro Vanoli
- Department of Molecular Medicine, University of Pavia
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, Viale Camillo, Pavia
| | - Barbara Cafferata
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo, Genova
| | | | - Claudia Mescoli
- Pathology Unit, Azienda Ospedaliera Padova, Via Ospedale Vecchio
| | - Mara Cananzi
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Via Ospedale Vecchio, Padova, Italy
| | - Rita Alaggio
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Roma
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Via Aristide Gabelli
- Veneto Institute of Oncology IOV-IRCCS, Viale Gattamelata, Padua
| | - Luca Mastracci
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
| | - Paola Francalanci
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Roma
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, San Giovanni Rotondo (FG), Italy
| |
Collapse
|
33
|
Sturm A, Atreya R, Bettenworth D, Bokemeyer B, Dignass A, Ehehalt R, Germer CT, Grunert PC, Helwig U, Horisberger K, Herrlinger K, Kienle P, Kucharzik T, Langhorst J, Maaser C, Ockenga J, Ott C, Siegmund B, Zeißig S, Stallmach A. Aktualisierte S3-Leitlinie „Diagnostik und Therapie des Morbus Crohn“ der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) (Version 4.1) – living guideline. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:1229-1318. [PMID: 39111333 DOI: 10.1055/a-2309-6123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Affiliation(s)
- Andreas Sturm
- Klinik für Innere Medizin mit Schwerpunkt Gastroenterologie, DRK Kliniken Berlin Westend, Berlin, Deutschland
| | - Raja Atreya
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Erlangen, Deutschland
| | | | - Bernd Bokemeyer
- Gastroenterologische Gemeinschaftspraxis Minden, Minden, Deutschland
| | - Axel Dignass
- Medizinische Klinik I, Agaplesion Markus Krankenhaus, Frankfurt am Main, Deutschland
| | | | | | - P C Grunert
- Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena, Deutschland
| | - Ulf Helwig
- Internistische Praxengemeinschaft, Oldenburg, Deutschland
| | - Karoline Horisberger
- Universitätsmedizin Johannes Gutenberg, Universität Klinik f. Allgemein-,Visceral- und Transplantationschirurgie, Mainz, Deutschland
| | | | - Peter Kienle
- Allgemein- und Viszeralchirurgie, Theresienkrankenhaus und Sankt Hedwig-Klinik GmbH, Mannheim, Deutschland
| | - Torsten Kucharzik
- Klinik für Allgemeine Innere Medizin und Gastroenterologie, Klinikum Lüneburg, Lüneburg, Deutschland
| | - Jost Langhorst
- Klinik für Integrative Medizin und Naturheilkunde, Klinikum am Bruderwald, Bamberg, Deutschland
| | - Christian Maaser
- Gastroenterologie, Ambulanzzentrum Lüneburg, Lüneburg, Deutschland
| | - Johann Ockenga
- Medizinische Klinik II, Klinikum Bremen Mitte - Gesundheit Nord, Bremen, Deutschland
| | - Claudia Ott
- Gastroenterologie Facharztzentrum, Regensburg, Deutschland
| | - Britta Siegmund
- Medizinische Klinik I, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Deutschland
| | - Sebastian Zeißig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Deutschland
| | - Andreas Stallmach
- Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena, Deutschland
| |
Collapse
|
34
|
Guz-Mark A, Aloi M, Scarallo L, Bramuzzo M, Escher JC, Alvisi P, Henderson P, Hojsak I, Lev-Tzion R, El-Matary W, Schwerd T, Granot M, Sladek M, Strisciuglio C, Müller KE, Olbjørn C, Tzivinikos C, Yerushalmy-Feler A, Huysentruyt K, Norsa L, Viola I, de Ridder L, Shouval DS, Lega S, Lionetti P, Catassi G, Assa A. Infantile and Very Early Onset Inflammatory Bowel Disease: A Multicenter Study. Pediatrics 2024; 154:e2023064546. [PMID: 39015095 DOI: 10.1542/peds.2023-064546] [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] [Received: 10/04/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVES This study described disease characteristics and long-term outcomes in patients diagnosed with very early onset inflammatory bowel disease (VEOIBD) (diagnosed before 6 years of age) and infantile-IBD (before 2 years). METHODS Cases from 21 centers worldwide diagnosed with VEOIBD (2008-2018), with minimum 2 years of follow-up, were retrospectively reviewed. RESULTS The cohort included 243 patients (52% males, median follow-up of 5.8 [range 2-18] years, including 69 [28%]) with infantile-IBD. IBD subtypes included Crohn's disease (CD), ulcerative colitis (UC), or IBD-unclassified (IBDU) in 30%, 59%, and 11%, respectively. Among patients with CD, 94% had colonic involvement, and among patients with UC/IBDU, 75% had pancolitis. Patients with infantile-IBD presented with higher rates of IBDU, lower hemoglobin and albumin levels, and higher C-reactive protein, and had lower response rates to first-induction therapy and corticosteroids therapy (P < .05 for all). Colectomy and diversion surgeries were performed in 11% and 4%, respectively, with no significant differences between age groups. Corticosteroid-free remission rates were 74% and 78% after 3 and 5 years, respectively, and 86% at end of follow-up. Genetic testing was performed in 96 (40%) patients. Among tested population, 15 (16%) were identified with monogenic disease. This group demonstrated lower response rates to induction therapies, higher rates of surgical intervention, and higher rates of major infections (P < .05 for all). CONCLUSIONS Patients with VEOIBD, including infantile-IBD, exhibit low rate of complications and surgical interventions at the long term. Patients with monogenic IBD are at risk for more severe disease course.
Collapse
Affiliation(s)
- Anat Guz-Mark
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Aloi
- Pediatric and Gastroenterology Unit, Sapienza University of Rome-Umberto I Hospital, Rome, Italy
| | - Luca Scarallo
- Gastroenterology and Nutrition Unit, Meyer children's Hospital, Florence, Italy
| | - Matteo Bramuzzo
- Gastroenterology, Digestive Endoscopy and Nutrition Unit, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo," Trieste, Italy
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Patrizia Alvisi
- Pediatric Gastroenterology unit, Maggiore Hospital, Bologna, Italy
| | - Paul Henderson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, Scotland
| | - Iva Hojsak
- Children's Hospital Zagreb, University of Zagreb Medical School, Zagreb, Croatia
| | - Raffi Lev-Tzion
- Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Wael El-Matary
- Pediatric Gastroenterology, University of Manitoba, Winnipeg, Canada
| | - Tobias Schwerd
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Maya Granot
- Pediatric Gastroenterology Unit, Edmond & Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Malgorzata Sladek
- Department of Pediatrics, Gastroenterology and Nutrition, Jagiellonian University Medical College, Krakow, Poland
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialistic Surgery, University of Campania Vanvitelli, Naples, Italy
| | - Katalin E Müller
- Heim Pal National Institute for Pediatrics, Budapest, Institute for Translational Medicine, University of Pécs, and Department of Family Care Methodology, Semmelweis University, Budapest, Hungary
| | - Christine Olbjørn
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Christos Tzivinikos
- Paediatric Gastroenterology Department, Al Jalila Children's Hospital, Mohammed Bin Rashid University, Dubai, United Arab Emirates
| | - Anat Yerushalmy-Feler
- Pediatric Gastroenterology Institute, "Dana-Dwek" Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Koen Huysentruyt
- Paediatric gastroenterology, KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lorenzo Norsa
- Pediatric Hepatology, Gastroenterology and Transplantation, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Irene Viola
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi," University of Messina, Italy
| | - Lissy de Ridder
- Department of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Dror S Shouval
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Tel-Aviv University, Tel-Aviv, Israel
| | - Sara Lega
- Gastroenterology, Digestive Endoscopy and Nutrition Unit, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo," Trieste, Italy
| | - Paolo Lionetti
- Department NEUROFARBA, Gastroenterology and Nutrition Unit, Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Giulia Catassi
- Pediatric and Gastroenterology Unit, Sapienza University of Rome-Umberto I Hospital, Rome, Italy
| | - Amit Assa
- Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
- The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
35
|
Lou Y, Lv Y, Yu J, Gu W, Jiang M, Chen J. MYO5B gene mutations may promote the occurrence of very early onset inflammatory bowel disease: a case report. BMC Med Genomics 2024; 17:187. [PMID: 39014344 PMCID: PMC11250955 DOI: 10.1186/s12920-024-01962-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND With recent advances in gene sequencing technology, more than 60 genetic mutations associated with very early onset inflammatory bowel disease (VEO-IBD) have been reported. Most of the genes are associated with immune deficiencies. The Myosin 5B (MYO5B) gene is primarily involved in cell motility and material transport which is associated with congenital intractable diarrhea and cholestasis. No studies have examined the relationship between the MYO5B gene and VEO-IBD. We report a case of a child with a mutation in the MYO5B gene who was diagnosed with VEO-IBD, then we investigated the association between the MYO5B gene and VEO-IBD. CASE PRESENTATION A 7-month-old baby girl with a chief complaint of "blood in the stool for more than 4 months and vaginal pus and blood discharge for 3 weeks" was diagnosed with VEO-IBD, and her symptoms improved after treatment with mesalazine. The whole-exome sequencing was performed with peripheral blood. Immunohistochemistry was performed on the terminal ileal tissue. Western blotting, quantitative polymerase chain reaction (Q-PCR) and immunofluorescence were performed with cultured organoid tissue from the terminal ileum. Whole-exome sequencing identified heterozygous missense of MYO5B variant of unknown significance (p. [I769N]; [T1546M]). Immunohistochemistry revealed a significant decrease in the expression of MYO5B protein in the terminal ileum of the child with MYO5B mutation; Q-PCR revealed a decrease in the mRNA levels of occludin and ZO-1 and both the mRNA levels and protein levels of MYO5B was downregulated in the patient. Immunofluorescence images showed that MYO5B gene mutation disrupted the apical delivery of transporters SGLT1, NHE3 and AQP7. CONCLUSIONS MYO5B gene mutation leading to the downregulation of MYO5B protein may promote the occurrence of VEO-IBD by decreasing mRNA and protein levels of intestinal tight junction genes and dislocating the apical transporters.
Collapse
Affiliation(s)
- Yue Lou
- Gastroenterology Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Rd, Bin Jiang District, Hangzhou, Zhejiang, 310052, China
| | - Yao Lv
- Gastroenterology Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Rd, Bin Jiang District, Hangzhou, Zhejiang, 310052, China
| | - Jindan Yu
- Gastroenterology Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Rd, Bin Jiang District, Hangzhou, Zhejiang, 310052, China
| | - Weizhong Gu
- Pathology Department, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Ming Jiang
- Gastroenterology Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Rd, Bin Jiang District, Hangzhou, Zhejiang, 310052, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, Zhejiang, China.
| | - Jie Chen
- Gastroenterology Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Rd, Bin Jiang District, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
36
|
Khavkin AI, Permyakova AA, Tsepilova MO, Kaplina AV, Sitkin SI, Surkov AN, Getmanov SD. Modern View on Very Early Onset and Early Onset Inflammatory Bowel Diseases in Children. CURRENT PEDIATRICS 2024; 23:145-151. [DOI: 10.15690/vsp.v23i3.2768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Nowadays, an urgent problem of pediatric gastroenterology is the study of inflammatory bowel diseases with very early onset (VEO-IBD), which have unique genetic, clinical, immunological, morphological, and laboratory sings. Early VEO-IBD is usually considered as monogenic disease, especially in combination with congenital immune defects, which leads to difficulties in diagnosis and management this pathology. Despite this, systematization of information about this group of nosological forms of IBD is practically not carried out. This article presents a review of the available information on etiological factors, course variants, and therapeutic options for VEO-IBD.
Collapse
Affiliation(s)
| | | | | | | | - Stanislav I. Sitkin
- Almazov National Medical Research Centre; North-Western State Medical University named after I.I. Mechnikov
| | - Andrey N. Surkov
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University
| | | |
Collapse
|
37
|
Ouahed JD, Griffith A, Collen LV, Snapper SB. Breaking Down Barriers: Epithelial Contributors to Monogenic IBD Pathogenesis. Inflamm Bowel Dis 2024; 30:1189-1206. [PMID: 38280053 PMCID: PMC11519031 DOI: 10.1093/ibd/izad319] [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: 09/22/2023] [Indexed: 01/29/2024]
Abstract
Monogenic causes of inflammatory bowel diseases (IBD) are increasingly being discovered. To date, much attention has been placed in those resulting from inborn errors of immunity. Therapeutic efforts have been largely focused on offering personalized immune modulation or curative bone marrow transplant for patients with IBD and underlying immune disorders. To date, less emphasis has been placed on monogenic causes of IBD that pertain to impairment of the intestinal epithelial barrier. Here, we provide a comprehensive review of monogenic causes of IBD that result in impaired intestinal epithelial barrier that are categorized into 6 important functions: (1) epithelial cell organization, (2) epithelial cell intrinsic functions, (3) epithelial cell apoptosis and necroptosis, (4) complement activation, (5) epithelial cell signaling, and (6) control of RNA degradation products. We illustrate how impairment of any of these categories can result in IBD. This work reviews the current understanding of the genes involved in maintaining the intestinal barrier, the inheritance patterns that result in dysfunction, features of IBD resulting from these disorders, and pertinent translational work in this field.
Collapse
Affiliation(s)
- Jodie D Ouahed
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexandra Griffith
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Lauren V Collen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Lerchova T, Størdal K, Andersson B, Ludvigsson J, Mårild K. Atopic Dermatitis in Early Childhood and Risk of Inflammatory Bowel Disease: A Scandinavian Birth Cohort Study. J Pediatr 2024; 270:114027. [PMID: 38521452 DOI: 10.1016/j.jpeds.2024.114027] [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: 02/05/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVE To examine the association between early-life atopic manifestations and later risk of inflammatory bowel disease (IBD), for which prospective data are scarce. STUDY DESIGN The population-based All Babies in Southeast Sweden (ABIS) and Norwegian Mother, Father, and Child (MoBa) cohorts follow children from birth (ABIS 1997-1999; MoBa 2000-2009) to the end of 2021. Based on validated questionnaires, parents prospectively reported information on asthma, food-related allergic symptoms, atopic dermatitis, and allergic rhinitis by age 3. IBD was defined by ≥ 2 diagnostic records in the national health registries. Cox regression estimated hazard ratios adjusted (aHRs) for parental IBD, atopy, education level, smoking habits, and national origin. Cohort-specific estimates were pooled using a random-effects model. RESULTS We compiled data on 83 311 children (ABIS, n = 9041; MoBa, n = 74 270). In over 1 174 756 person-years of follow-up, 301 participants were diagnosed with IBD. Children with atopic dermatitis at age 3 had an increased risk of IBD (pooled aHR = 1.46 [95% CI = 1.13-1.88]), Crohn's disease (pooled aHR = 1.53 [95%CI = 1.04-2.26]), and ulcerative colitis (pooled aHR = 1.78 [95%CI = 1.15-2.75]). Conversely, any atopic manifestation by age 3 was not associated with IBD (pooled aHR = 1.20 [95%CI = 0.95-1.52]), nor were analyses specifically focused on early-life food-related allergic symptoms, asthma, and allergic rhinitis. CONCLUSION While atopic manifestations in early childhood were overall not associated with IBD, children with atopic dermatitis specifically were at increased risk of developing IBD, suggesting shared etiologic traits; these findings might be useful in identifying at-risk individuals for IBD.
Collapse
Affiliation(s)
- Tereza Lerchova
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Ketil Størdal
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway; Children's Center, Oslo University Hospital, Oslo, Norway
| | - Björn Andersson
- Bioinformatics and Data Centre, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Linköping, Sweden; Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Karl Mårild
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, Queen Silvia Children's Hospital, Gothenburg, Sweden
| |
Collapse
|
39
|
Baba A, Aregbesola A, Caldwell PHY, Elliott SA, Elsman EBM, Fernandes RM, Hartling L, Heath A, Kelly LE, Preston J, Sammy A, Webbe J, Williams K, Woolfall K, Klassen TP, Offringa M. Developments in the Design, Conduct, and Reporting of Child Health Trials. Pediatrics 2024; 154:e2024065799. [PMID: 38832441 DOI: 10.1542/peds.2024-065799] [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] [Received: 01/17/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 06/05/2024] Open
Abstract
To identify priority areas to improve the design, conduct, and reporting of pediatric clinical trials, the international expert network, Standards for Research (StaR) in Child Health, was assembled and published the first 6 Standards in Pediatrics in 2012. After a recent review summarizing the 247 publications by StaR Child Health authors that highlight research practices that add value and reduce research "waste," the current review assesses the progress in key child health trial methods areas: consent and recruitment, containing risk of bias, roles of data monitoring committees, appropriate sample size calculations, outcome selection and measurement, and age groups for pediatric trials. Although meaningful change has occurred within the child health research ecosystem, measurable progress is still disappointingly slow. In this context, we identify and review emerging trends that will advance the agenda of increased clinical usefulness of pediatric trials, including patient and public engagement, Bayesian statistical approaches, adaptive designs, and platform trials. We explore how implementation science approaches could be applied to effect measurable improvements in the design, conducted, and reporting of child health research.
Collapse
Affiliation(s)
- Ami Baba
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Alex Aregbesola
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Patrina H Y Caldwell
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia
| | - Sarah A Elliott
- Cochrane Child Health
- Alberta Research Centre for Health Evidence, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ellen B M Elsman
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Ricardo M Fernandes
- Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Lisa Hartling
- Cochrane Child Health
- Alberta Research Centre for Health Evidence, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anna Heath
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Statistical Science, University College London, London, United Kingdom
| | - Lauren E Kelly
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pharmacology and Therapeutics, Rady Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Jennifer Preston
- National Institute for Health and Care Research (NIHR) Alder Hey Clinical Research Facility, Liverpool, United Kingdom
| | - Adrian Sammy
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - James Webbe
- Section of Neonatal Medicine, Imperial College London, London, United Kingdom
| | - Katrina Williams
- Department of Paediatrics, Monash University and Developmental Paediatrics, Monash Children's Hospital, Melbourne, Australia
| | - Kerry Woolfall
- Department of Public Health, Policy and Systems, University of Liverpool, Liverpool, United Kingdom
| | - Terry P Klassen
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Martin Offringa
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Eslamian G, Jamee M, Momen T, Rohani P, Ebrahimi S, Mesdaghi M, Ghadimi S, Mansouri M, Mahdaviani SA, Sadeghi-shabestari M, Fallahpour M, Shamsian BS, Eslami N, Sharafian S, Dara N, Nasri P, Amini N, Enayat J, Fallahi M, Ghasemi Hashtrodi L, Shojaei M, Guevara Becerra M, Uhlig HH, Chavoshzadeh Z. Genomic testing identifies monogenic causes in patients with very early-onset inflammatory bowel disease: a multicenter survey in an Iranian cohort. Clin Exp Immunol 2024; 217:1-11. [PMID: 38651248 PMCID: PMC11188541 DOI: 10.1093/cei/uxae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/15/2024] [Accepted: 04/20/2024] [Indexed: 04/25/2024] Open
Abstract
Patients with very early-onset inflammatory bowel disease (VEO-IBD) may present because of underlying monogenic inborn errors of immunity (IEI). Strong differences have been observed in the causes of monogenic IBD among ethnic populations. This multicenter study was carried out on 16 Iranian patients with VEO-IBD. We reviewed clinical and basic immunologic evaluation including flow cytometry and immunoglobulin levels. All patients underwent clinical whole exome sequencing (WES). Sixteen patients (8 females and 8 males) with a median age of 43.5 months were enrolled. The median age at the onset of symptoms was 4 months. Most patients (12, 75%) had consanguineous parents. Chronic non-bloody diarrhea (13, 81.3%) and perianal diseases including perianal abscess (6, 37.5%), anal fissure (6, 37.5%), or anal fistula (2, 12.5%) were the most common manifestations. WES identified a spectrum of genetic variants in 13 patients (81.3%): IL10RB (6, 37.5%), MVK (3, 18.8%), and CASP8, SLC35C1, G6PC3, and IKBKB in 1 patient, respectively. In 3 patients (18.7%), no variant was identified. Flow cytometry identified a spectrum of abnormalities that helped to assess the evidence of genetic diagnosis. At the end of the survey, 3 (18.8%) patients were deceased. This high rate of monogenic defects with a broad spectrum of genes reiterates the importance of investigating IEI in patients with infantile-onset IBD.
Collapse
Affiliation(s)
- Golnaz Eslamian
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Jamee
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pediatric Nephrology Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tooba Momen
- Department of Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pejman Rohani
- Pediatric Gastroenterology and Hepatology Research Center, Children’s Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mehrnaz Mesdaghi
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soodeh Ghadimi
- School of Medicine, Azad University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Mansouri
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Mahdaviani
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Sadeghi-shabestari
- Immunology Research Center, TB and Lung Research Center, Children Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Fallahpour
- Allergy Department, Rasoul Akram Complex, Clinical Research Development Center (RCRDC), Iran University of Medical Sciences, Tehran, Iran
| | - Bibi Shahin Shamsian
- Pediatric Congenital Hematologic Disorders Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Eslami
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samin Sharafian
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naghi Dara
- Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peiman Nasri
- Department of Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Metabolic Liver Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Niloufar Amini
- Department of Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javad Enayat
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mazdak Fallahi
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Ghasemi Hashtrodi
- Children Growth Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Ghods Hospital, Qazvin, Iran
| | - Mohammad Shojaei
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Martha Guevara Becerra
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Zahra Chavoshzadeh
- Immunology and Allergy Department, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Tóbi L, Prehoda B, Balogh AM, Nagypál P, Kovács K, Miheller P, Iliás Á, Dezsőfi-Gottl A, Cseh Á. Transition is associated with lower disease activity, fewer relapses, better medication adherence, and lower lost-to-follow-up rate as opposed to self-transfer in pediatric-onset inflammatory bowel disease patients: results of a longitudinal, follow-up, controlled study. Therap Adv Gastroenterol 2024; 17:17562848241252947. [PMID: 39156978 PMCID: PMC11327998 DOI: 10.1177/17562848241252947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/15/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Despite the continuously rising rate of pediatric-onset inflammatory bowel diseases (PIBD), there are no consensus transitional guidelines or standardized practices. OBJECTIVES We aimed to examine: (1) the determinants of a successful transfer, (2) the effects of the transfer versus transition on the disease course and patient compliance, (3) the unique characteristics of PIBD patients, that need special attention in adult care. DESIGN Longitudinal, follow-up, controlled study conducted between 2001 and 2022, with retrospective data collection until 2018, thence prospective. METHODS Three hundred fifty-one PIBD patients enrolled in the study, of whom 152 were moved to adult care, with a mean post-transfer follow-up time of 3 years. Seventy-three patients took part in structured transition, whereas 79 self-transferred to adult care. The main outcome measures were disease activity (defined by PCDAI, PUCAI, CDAI, and Mayo-scores) and course, hospitalizations, surgeries, IBD-related complications, including anthropometry and bone density, patient compliance, medication adherence, and continuation of medical care. RESULTS Patients who underwent structured transition spent significantly more time in remission (83.6% ± 28.5% versus 77.5% ± 29.7%, p = 0.0339) and had better adherence to their medications (31.9% versus 16.4% non-adherence rate, p = 0.0455) in adult care, with self-transferred patients having a 1.59-fold increased risk of discontinuing their medical care and a 1.88-fold increased risk of experiencing a relapse. Post-transfer the compliance of patients deteriorated (38.5% versus 29%, p = 0.0002), with the highest lost-to-follow-up rate during the changing period between the healthcare systems (12.7%), in which female gender was a risk factor (p = 0.010). PIBD patients had experienced IBD-related complications (23.4%) and former surgeries (15%) upon arriving at adult care, with high rates of malnutrition, growth impairment, and poor bone health. CONCLUSION Structured transition plays a key role in ensuring the best disease course and lowering the lost-to-follow-up rate among PIBD patients. BRIEF SUMMARY Structured transition plays a key role in ensuring the best disease outcome among PIBD patients, as in our study it was associated with lower disease activity, fewer relapses, better medication adherence, and lower lost-to-follow-up rate as opposed to self-transfer.
Collapse
Affiliation(s)
- Luca Tóbi
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Post Office Box 2, Budapest 1428, Hungary
| | - Bence Prehoda
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Anna M. Balogh
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Petra Nagypál
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Krisztián Kovács
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Pál Miheller
- Department of Surgery, Transplantation, and Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Ákos Iliás
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Antal Dezsőfi-Gottl
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Áron Cseh
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| |
Collapse
|
42
|
Mao Z, Betti MJ, Cedeno MA, Pedroza LA, Basaria S, Liu Q, Choi JM, Markle JG. Clinical and cellular phenotypes resulting from a founder mutation in IL10RB. Clin Exp Immunol 2024; 216:113-119. [PMID: 37503744 PMCID: PMC11036105 DOI: 10.1093/cei/uxad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/27/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023] Open
Abstract
Inborn errors of immunity are a group of rare genetically determined diseases that impair immune system development or function. Many of these diseases include immune dysregulation, autoimmunity, or autoinflammation as prominent clinical features. In some children diagnosed with very early onset inflammatory bowel disease (VEOIBD), monogenic inborn errors of immune dysregulation underlie disease. We report a case of VEOIBD caused by a novel homozygous loss of function mutation in IL10RB. We use cytometry by time-of-flight with a broad panel of antibodies to interrogate the immunophenotype of this patient and detect reduced frequencies of CD4 and CD8 T cells with additional defects in some populations of T helper cells, innate-like T cells, and memory B cells. Finally, we identify the patient's mutation as a founder allele in an isolated indigenous population and estimate the age of this variant by studying the shared ancestral haplotype.
Collapse
Affiliation(s)
- Zhiming Mao
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael J Betti
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Miguel A Cedeno
- Department of Pediatrics, Hospital de ninos Roberto Gilbert Elizalde, Guayaquil, Ecuador
| | - Luis A Pedroza
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Shamel Basaria
- Division of Molecular Pathogenesis, Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qi Liu
- Division of Molecular Pathogenesis, Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph M Choi
- Division of Molecular Pathogenesis, Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Janet G Markle
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Molecular Pathogenesis, Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
43
|
Marafini I, Monteleone I, Laudisi F, Monteleone G. Aryl Hydrocarbon Receptor Signalling in the Control of Gut Inflammation. Int J Mol Sci 2024; 25:4527. [PMID: 38674118 PMCID: PMC11050475 DOI: 10.3390/ijms25084527] [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: 03/25/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Aryl hydrocarbon receptor (AHR), a transcription factor activated by many natural and synthetic ligands, represents an important mediator of the interplay between the environment and the host's immune responses. In a healthy gut, AHR activation promotes tolerogenic signals, which help maintain mucosal homeostasis. AHR expression is defective in the inflamed gut of patients with inflammatory bowel diseases (IBD), where decreased AHR signaling is supposed to contribute to amplifying the gut tissue's destructive immune-inflammatory responses. We here review the evidence supporting the role of AHR in controlling the "physiological" intestinal inflammation and summarize the data about the therapeutic effects of AHR activators, both in preclinical mouse models of colitis and in patients with IBD.
Collapse
Affiliation(s)
- Irene Marafini
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
| | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of “Tor Vergata”, 00133 Rome, Italy;
| | - Federica Laudisi
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy;
| | - Giovanni Monteleone
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy;
| |
Collapse
|
44
|
Abstract
Paediatric-onset inflammatory bowel disease (IBD) is a complex and heterogenous condition. Incidence of disease in those aged <18 years has doubled over the last 25 years, with concurrent increased prevalence and no decrease in disease severity. The tools available at diagnosis for investigation have developed over the last 10 years, including better utilisation of faecal calprotectin, improved small bowel imaging and video capsule endoscopy. Alongside this, management options have increased and include biological and small molecule therapies targeting alternative pathways (such as interleukin 12/23, integrins and Janus kinase/signal transducers and activators of transcription, JAK-STAT pathways) and better understanding of therapeutic drug monitoring for more established agents, such as infliximab. Dietary manipulation remains an interesting but contentious topic.This review summarises some of the recent developments in the diagnosis, investigation and management of IBD in children and young people. IBD is increasingly recognised as a continuum of disease, with a proportion of patients presenting with classical Crohn's disease or ulcerative colitis phenotypes. Future implementation of personalisation and stratification strategies, including clinical and molecular biomarkers, implementation of predictors of response and outcome and use of additional therapies, will continue to require working within clinical networks and multiprofessional teams.
Collapse
Affiliation(s)
- James John Ashton
- Department of Paediatric Gastroenterology, Southampton Children's Hospital, Southampton, UK
- Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - R Mark Beattie
- Department of Paediatric Gastroenterology, Southampton Children's Hospital, Southampton, UK
| |
Collapse
|
45
|
Diamanti A, Trovato CM, Gandullia P, Lezo A, Spagnuolo MI, Bolasco G, Capriati T, Lacitignola L, Norsa L, Francalanci P, Novelli A. Intractable diarrhea in infancy and molecular analysis: We are beyond the tip of the iceberg. Dig Liver Dis 2024; 56:607-612. [PMID: 38044226 DOI: 10.1016/j.dld.2023.09.002] [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: 03/28/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Intractable diarrhea (ID) could be defined as a syndrome of severe chronic diarrhea associated with malnutrition not easily resolved by conventional management. AIMS To provide an overview on etiology and management of ID patients in Italy in the last 12 years. METHODS The members of Italian Society for Pediatric Gastroenterology, Hepatology and Nutrition (SIGENP) enrolled all ID patients seen between January 1, 2011 and December 31, 2022. RESULTS 69 children were enrolled (49 M, 20 F; median age at ID onset 9.5 days) from 7 tertiary care pediatric centers. Overall 62 patients had genetic diseases; 3 had infantile Inflammatory Bowel Disease and 1 autoimmune enteropathy in absence of genetic mutations; 2 undefined ID. Defects of intestinal immune-related homeostasis caused ID in 29 patients (42 %). CONCLUSION ID is a rare but challenging problem, although the potential for diagnosis has improved over time. In particular, molecular analysis allowed to identity genetic defects in 90 % of patients and to detect new genetic mutations responsible for ID. Due to both the challenging diagnosis and the treatment for many of these diseases, the close relationship between immune system and digestive tract should require a close collaboration between pediatric immunologists and gastroenterologists, to optimize epidemiologic surveillance and management of ID.
Collapse
Affiliation(s)
- A Diamanti
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy.
| | - C M Trovato
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - P Gandullia
- Pediatric Gastroenterology and Endoscopy, IRCCS G. Gaslini Institute, via Gerolamo Gaslini 5, Genoa, Italy
| | - A Lezo
- Dietetics and Clinical Nutrition Unit, Children's Hospital Regina Margherita, Città della Salute e della Scienza Torino, Turin, Italy
| | - M I Spagnuolo
- Department of Translation Medical Science, Pediatric Section, University Federico II, Naples, Italy
| | - G Bolasco
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - T Capriati
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - L Lacitignola
- Department NEUROFARBA, University of Florence. Meyer Children's Hospital, viale Gaetano Pieraccini 24, Florence, Italy
| | - L Norsa
- Regional Health Care and Social Agency Papa Giovanni XXIII, The Netherlands
| | - P Francalanci
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - A Novelli
- Hepatology, Gastroenterology and Nutrition Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
46
|
Hall CHT, de Zoeten EF. Understanding very early onset inflammatory bowel disease (VEOIBD) in relation to inborn errors of immunity. Immunol Rev 2024; 322:329-338. [PMID: 38115672 PMCID: PMC11044353 DOI: 10.1111/imr.13302] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023]
Abstract
Inflammatory bowel diseases (IBD) are multifactorial diseases which are caused by the combination of genetic predisposition, exposure factors (environmental and dietary), immune status, and dysbiosis. IBD is a disease which presents at any age, ranging from newborns to the elderly. The youngest of the pediatric IBD population have a more unique presentation and clinical course and may have a different etiology. Very early onset IBD (VEOIBD) patients, designated as those diagnosed prior the age of 6, have distinct features which are more frequent in this patient population including increased incidence of monogenetic causes for IBD (0%-33% depending on the study). This proportion is increased in the youngest subsets, which is diagnosed prior to the age of 2. To date, there are approximately 80 monogenic causes of VEOIBD that have been identified and published. Many of these monogenic causes are inborn errors of immunity yet the majority of VEOIBD patients do not have an identifiable genetic cause for their disease. In this review, we will focus on the clinical presentation, evaluation, and monogenic categories which have been associated with VEOIBD including (1) Epithelial cell defects (2) Adaptive immune defects, (3) Innate Immune/Bacterial Clearance and Recognition defects, and (4) Hyperinflammatory and autoinflammatory disorders. We will highlight differential diagnosis of VEOIBD presentations, as well as evaluation and treatment, which will be helpful for those who study and care for VEOIBD patients outside of the pediatric gastroenterology field. This is a fast-moving field of research which has grown significantly based on knowledge that we gain from our patients. These scientific findings have identified novel mucosal biology pathways and will continue to inform our understanding of gastrointestinal biology.
Collapse
Affiliation(s)
- Caroline H. T. Hall
- Mucosal Inflammation Program University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edwin F. de Zoeten
- Mucosal Inflammation Program University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
47
|
Chang JY, Park SJ, Park JJ, Kim TI, Cheon JH, Park J. Impact of age at diagnosis on long-term prognosis in patients with intestinal Behçet's disease. J Gastroenterol Hepatol 2024; 39:519-526. [PMID: 38149352 DOI: 10.1111/jgh.16449] [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: 10/02/2023] [Revised: 11/22/2023] [Accepted: 12/03/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND AND AIM Although age at disease onset is considered to be a significant factor in the prognosis of Crohn's disease, little is known about its influence on the long-term prognosis of those with intestinal Behçet's disease (BD). This study aimed to evaluate the long-term clinical outcomes of patients with intestinal BD according to age of disease onset. METHODS Patients diagnosed with intestinal BD at < 18, 18-60, and > 60 years of age were classified into early-onset, adult-onset, and late-onset groups, respectively. The influence of disease onset time on clinical prognosis, including specific medical requirements, BD-related intestinal surgery, hospitalization, and emergency room visits, was compared using the log-rank test in a large cohort of patients with intestinal BD. RESULTS Among 780 patients, 21 (2.7%), 672 (86.2%), and 87 (11.1%) comprised the early-onset, adult-onset, and late-onset groups, respectively. Patients in the early-onset group were more likely to require immunosuppressants than those in the adult-onset group (P = 0.048). Nine (42.9%), 158 (23.5%), and 18 (20.7%) patients in the early-onset, adult-onset, and late-onset groups, respectively, underwent intestinal resection. The early-onset group exhibited a higher risk for intestinal resection than the late-onset (P = 0.043) and adult-onset (P = 0.030) groups. The late-onset group exhibited a higher risk for BD-related hospitalization than the adult-onset group (P = 0.023). CONCLUSIONS Age at diagnosis affected the clinical course of intestinal BD, including intestinal surgery, hospitalization, and specific medical requirements. Different treatment strategies should be established according to age at diagnosis.
Collapse
Affiliation(s)
- Ji Young Chang
- Department of Health Promotion Medicine, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Soo Jung Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Inflammatory Bowel Disease Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Jun Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Inflammatory Bowel Disease Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Il Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Inflammatory Bowel Disease Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Hee Cheon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Inflammatory Bowel Disease Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jihye Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Inflammatory Bowel Disease Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
48
|
O'Brien CL, Summers KM, Martin NM, Carter-Cusack D, Yang Y, Barua R, Dixit OVA, Hume DA, Pavli P. The relationship between extreme inter-individual variation in macrophage gene expression and genetic susceptibility to inflammatory bowel disease. Hum Genet 2024; 143:233-261. [PMID: 38421405 PMCID: PMC11043138 DOI: 10.1007/s00439-024-02642-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/14/2024] [Indexed: 03/02/2024]
Abstract
The differentiation of resident intestinal macrophages from blood monocytes depends upon signals from the macrophage colony-stimulating factor receptor (CSF1R). Analysis of genome-wide association studies (GWAS) indicates that dysregulation of macrophage differentiation and response to microorganisms contributes to susceptibility to chronic inflammatory bowel disease (IBD). Here, we analyzed transcriptomic variation in monocyte-derived macrophages (MDM) from affected and unaffected sib pairs/trios from 22 IBD families and 6 healthy controls. Transcriptional network analysis of the data revealed no overall or inter-sib distinction between affected and unaffected individuals in basal gene expression or the temporal response to lipopolysaccharide (LPS). However, the basal or LPS-inducible expression of individual genes varied independently by as much as 100-fold between subjects. Extreme independent variation in the expression of pairs of HLA-associated transcripts (HLA-B/C, HLA-A/F and HLA-DRB1/DRB5) in macrophages was associated with HLA genotype. Correlation analysis indicated the downstream impacts of variation in the immediate early response to LPS. For example, variation in early expression of IL1B was significantly associated with local SNV genotype and with subsequent peak expression of target genes including IL23A, CXCL1, CXCL3, CXCL8 and NLRP3. Similarly, variation in early IFNB1 expression was correlated with subsequent expression of IFN target genes. Our results support the view that gene-specific dysregulation in macrophage adaptation to the intestinal milieu is associated with genetic susceptibility to IBD.
Collapse
Affiliation(s)
- Claire L O'Brien
- Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
- Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia
| | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Natalia M Martin
- Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia
| | - Dylan Carter-Cusack
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Yuanhao Yang
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Rasel Barua
- Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia
| | - Ojas V A Dixit
- Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
| | - Paul Pavli
- Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia.
- School of Medicine and Psychology, College of Health and Medicine, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
49
|
The Critical Assessment of Genome Interpretation Consortium, Jain S, Bakolitsa C, Brenner SE, Radivojac P, Moult J, Repo S, Hoskins RA, Andreoletti G, Barsky D, Chellapan A, Chu H, Dabbiru N, Kollipara NK, Ly M, Neumann AJ, Pal LR, Odell E, Pandey G, Peters-Petrulewicz RC, Srinivasan R, Yee SF, Yeleswarapu SJ, Zuhl M, Adebali O, Patra A, Beer MA, Hosur R, Peng J, Bernard BM, Berry M, Dong S, Boyle AP, Adhikari A, Chen J, Hu Z, Wang R, Wang Y, Miller M, Wang Y, Bromberg Y, Turina P, Capriotti E, Han JJ, Ozturk K, Carter H, Babbi G, Bovo S, Di Lena P, Martelli PL, Savojardo C, Casadio R, Cline MS, De Baets G, Bonache S, Díez O, Gutiérrez-Enríquez S, Fernández A, Montalban G, Ootes L, Özkan S, Padilla N, Riera C, De la Cruz X, Diekhans M, Huwe PJ, Wei Q, Xu Q, Dunbrack RL, Gotea V, Elnitski L, Margolin G, Fariselli P, Kulakovskiy IV, Makeev VJ, Penzar DD, Vorontsov IE, Favorov AV, Forman JR, Hasenahuer M, Fornasari MS, Parisi G, Avsec Z, Çelik MH, Nguyen TYD, Gagneur J, Shi FY, Edwards MD, Guo Y, Tian K, Zeng H, Gifford DK, Göke J, Zaucha J, Gough J, Ritchie GRS, Frankish A, Mudge JM, Harrow J, Young EL, et alThe Critical Assessment of Genome Interpretation Consortium, Jain S, Bakolitsa C, Brenner SE, Radivojac P, Moult J, Repo S, Hoskins RA, Andreoletti G, Barsky D, Chellapan A, Chu H, Dabbiru N, Kollipara NK, Ly M, Neumann AJ, Pal LR, Odell E, Pandey G, Peters-Petrulewicz RC, Srinivasan R, Yee SF, Yeleswarapu SJ, Zuhl M, Adebali O, Patra A, Beer MA, Hosur R, Peng J, Bernard BM, Berry M, Dong S, Boyle AP, Adhikari A, Chen J, Hu Z, Wang R, Wang Y, Miller M, Wang Y, Bromberg Y, Turina P, Capriotti E, Han JJ, Ozturk K, Carter H, Babbi G, Bovo S, Di Lena P, Martelli PL, Savojardo C, Casadio R, Cline MS, De Baets G, Bonache S, Díez O, Gutiérrez-Enríquez S, Fernández A, Montalban G, Ootes L, Özkan S, Padilla N, Riera C, De la Cruz X, Diekhans M, Huwe PJ, Wei Q, Xu Q, Dunbrack RL, Gotea V, Elnitski L, Margolin G, Fariselli P, Kulakovskiy IV, Makeev VJ, Penzar DD, Vorontsov IE, Favorov AV, Forman JR, Hasenahuer M, Fornasari MS, Parisi G, Avsec Z, Çelik MH, Nguyen TYD, Gagneur J, Shi FY, Edwards MD, Guo Y, Tian K, Zeng H, Gifford DK, Göke J, Zaucha J, Gough J, Ritchie GRS, Frankish A, Mudge JM, Harrow J, Young EL, Yu Y, Huff CD, Murakami K, Nagai Y, Imanishi T, Mungall CJ, Jacobsen JOB, Kim D, Jeong CS, Jones DT, Li MJ, Guthrie VB, Bhattacharya R, Chen YC, Douville C, Fan J, Kim D, Masica D, Niknafs N, Sengupta S, Tokheim C, Turner TN, Yeo HTG, Karchin R, Shin S, Welch R, Keles S, Li Y, Kellis M, Corbi-Verge C, Strokach AV, Kim PM, Klein TE, Mohan R, Sinnott-Armstrong NA, Wainberg M, Kundaje A, Gonzaludo N, Mak ACY, Chhibber A, Lam HYK, Dahary D, Fishilevich S, Lancet D, Lee I, Bachman B, Katsonis P, Lua RC, Wilson SJ, Lichtarge O, Bhat RR, Sundaram L, Viswanath V, Bellazzi R, Nicora G, Rizzo E, Limongelli I, Mezlini AM, Chang R, Kim S, Lai C, O’Connor R, Topper S, van den Akker J, Zhou AY, Zimmer AD, Mishne G, Bergquist TR, Breese MR, Guerrero RF, Jiang Y, Kiga N, Li B, Mort M, Pagel KA, Pejaver V, Stamboulian MH, Thusberg J, Mooney SD, Teerakulkittipong N, Cao C, Kundu K, Yin Y, Yu CH, Kleyman M, Lin CF, Stackpole M, Mount SM, Eraslan G, Mueller NS, Naito T, Rao AR, Azaria JR, Brodie A, Ofran Y, Garg A, Pal D, Hawkins-Hooker A, Kenlay H, Reid J, Mucaki EJ, Rogan PK, Schwarz JM, Searls DB, Lee GR, Seok C, Krämer A, Shah S, Huang CV, Kirsch JF, Shatsky M, Cao Y, Chen H, Karimi M, Moronfoye O, Sun Y, Shen Y, Shigeta R, Ford CT, Nodzak C, Uppal A, Shi X, Joseph T, Kotte S, Rana S, Rao A, Saipradeep VG, Sivadasan N, Sunderam U, Stanke M, Su A, Adzhubey I, Jordan DM, Sunyaev S, Rousseau F, Schymkowitz J, Van Durme J, Tavtigian SV, Carraro M, Giollo M, Tosatto SCE, Adato O, Carmel L, Cohen NE, Fenesh T, Holtzer T, Juven-Gershon T, Unger R, Niroula A, Olatubosun A, Väliaho J, Yang Y, Vihinen M, Wahl ME, Chang B, Chong KC, Hu I, Sun R, Wu WKK, Xia X, Zee BC, Wang MH, Wang M, Wu C, Lu Y, Chen K, Yang Y, Yates CM, Kreimer A, Yan Z, Yosef N, Zhao H, Wei Z, Yao Z, Zhou F, Folkman L, Zhou Y, Daneshjou R, Altman RB, Inoue F, Ahituv N, Arkin AP, Lovisa F, Bonvini P, Bowdin S, Gianni S, Mantuano E, Minicozzi V, Novak L, Pasquo A, Pastore A, Petrosino M, Puglisi R, Toto A, Veneziano L, Chiaraluce R, Ball MP, Bobe JR, Church GM, Consalvi V, Cooper DN, Buckley BA, Sheridan MB, Cutting GR, Scaini MC, Cygan KJ, Fredericks AM, Glidden DT, Neil C, Rhine CL, Fairbrother WG, Alontaga AY, Fenton AW, Matreyek KA, Starita LM, Fowler DM, Löscher BS, Franke A, Adamson SI, Graveley BR, Gray JW, Malloy MJ, Kane JP, Kousi M, Katsanis N, Schubach M, Kircher M, Mak ACY, Tang PLF, Kwok PY, Lathrop RH, Clark WT, Yu GK, LeBowitz JH, Benedicenti F, Bettella E, Bigoni S, Cesca F, Mammi I, Marino-Buslje C, Milani D, Peron A, Polli R, Sartori S, Stanzial F, Toldo I, Turolla L, Aspromonte MC, Bellini M, Leonardi E, Liu X, Marshall C, McCombie WR, Elefanti L, Menin C, Meyn MS, Murgia A, Nadeau KCY, Neuhausen SL, Nussbaum RL, Pirooznia M, Potash JB, Dimster-Denk DF, Rine JD, Sanford JR, Snyder M, Cote AG, Sun S, Verby MW, Weile J, Roth FP, Tewhey R, Sabeti PC, Campagna J, Refaat MM, Wojciak J, Grubb S, Schmitt N, Shendure J, Spurdle AB, Stavropoulos DJ, Walton NA, Zandi PP, Ziv E, Burke W, Chen F, Carr LR, Martinez S, Paik J, Harris-Wai J, Yarborough M, Fullerton SM, Koenig BA, McInnes G, Shigaki D, Chandonia JM, Furutsuki M, Kasak L, Yu C, Chen R, Friedberg I, Getz GA, Cong Q, Kinch LN, Zhang J, Grishin NV, Voskanian A, Kann MG, Tran E, Ioannidis NM, Hunter JM, Udani R, Cai B, Morgan AA, Sokolov A, Stuart JM, Minervini G, Monzon AM, Batzoglou S, Butte AJ, Greenblatt MS, Hart RK, Hernandez R, Hubbard TJP, Kahn S, O’Donnell-Luria A, Ng PC, Shon J, Veltman J, Zook JM. CAGI, the Critical Assessment of Genome Interpretation, establishes progress and prospects for computational genetic variant interpretation methods. Genome Biol 2024; 25:53. [PMID: 38389099 PMCID: PMC10882881 DOI: 10.1186/s13059-023-03113-6] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 11/17/2023] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND The Critical Assessment of Genome Interpretation (CAGI) aims to advance the state-of-the-art for computational prediction of genetic variant impact, particularly where relevant to disease. The five complete editions of the CAGI community experiment comprised 50 challenges, in which participants made blind predictions of phenotypes from genetic data, and these were evaluated by independent assessors. RESULTS Performance was particularly strong for clinical pathogenic variants, including some difficult-to-diagnose cases, and extends to interpretation of cancer-related variants. Missense variant interpretation methods were able to estimate biochemical effects with increasing accuracy. Assessment of methods for regulatory variants and complex trait disease risk was less definitive and indicates performance potentially suitable for auxiliary use in the clinic. CONCLUSIONS Results show that while current methods are imperfect, they have major utility for research and clinical applications. Emerging methods and increasingly large, robust datasets for training and assessment promise further progress ahead.
Collapse
|
50
|
Amaryan G, Sarkisian T, Tadevosyan A, Braegger C. Familial Mediterranean fever in Armenian children with inflammatory bowel disease. Front Pediatr 2024; 11:1288523. [PMID: 38410520 PMCID: PMC10895960 DOI: 10.3389/fped.2023.1288523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/27/2023] [Indexed: 02/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) and familial Mediterranean fever (FMF) are inflammatory diseases with complex interactions among genetic, immune, and environmental factors. FMF is a monogenic autoinflammatory disease, characterized by recurrent febrile attacks and polyserositis, and is manifested mainly in childhood. FMF is widespread in Armenia. There are reports on the concurrent occurrence of FMF and IBD. MEFV gene mutations may have a disease-modifying effect on IBD. We have investigated the frequency of MEFV mutations and FMF in Armenian children with IBD and their influence on the clinical course. A total of 69 untreated IBD patients under 18 years of age were enrolled: 52.1% (36) had ulcerative colitis (UC), 21.7% (15) had Crohn's disease (CD), and 26.0% (18) had unclassified colitis (IBD-U). The frequency of FMF among them was 36.2% (25/69), and MEFV mutations were identified in 53.6% (37/69). The highest rate of MEFV mutations and FMF was in UC patients (61.1% and 41.6% respectively). In all, 56.7% (21/37) of IBD patients with MEFV mutations had M694V mutated alleles, mainly in compound heterozygous and heterozygous states. There were no associations in the group of IBD patients with coexisting FMF (25), either between any MEFV mutation and type of IBD or coexistence of FMF. Overall, 36.0% (9/25) of them developed VEO IBD and carried mainly the M694V mutation. We concluded that the carrier frequency of MEFV mutations among Armenian pediatric IBD patients was rather high (53.6%), especially for UC. It was suggested that the MEFV gene is not necessarily a susceptibility gene but most likely modifies the course of IBD. MEFV genetic testing was recommended for Armenian pediatric IBD patients, especially for VEO UC and IBD-U, atypical IBD course, or resistance to the conventional treatment. They should also be asked for isolated febrile attacks, recurrent arthritis, and family history, even in the absence of FMF typical symptoms, to rule out FMF and its complications.
Collapse
Affiliation(s)
- Gayane Amaryan
- National Pediatrics Center for Familial Mediterranean Fever, “Arabkir” Medical Complex-Institute of Child and Adolescent Health, Yerevan, Armenia
- Department of Pediatrics, Yerevan State Medical University, Yerevan, Armenia
| | - Tamara Sarkisian
- Center of Medical Genetics and Primary Health Care, Yerevan, Armenia
- Department of Medical Genetics, Yerevan State Medical University, Yerevan, Armenia
| | - Artashes Tadevosyan
- Department of Public Health and Health Care Organization, Yerevan State Medical University, Yerevan, Armenia
| | - Christian Braegger
- Nutrition Research Unit, Children’s University Hospital in Zürich, Zürich, Switzerland
| |
Collapse
|