1
|
Coburn S, Trojanowski PJ, Vagadori J, Hinds P, Germone M, Liu E, Streisand R, Bost J. Development and Validation of a Pediatric Celiac Disease-Specific Quality of Life Measure. Am J Gastroenterol 2025; 120:1116-1126. [PMID: 39401060 PMCID: PMC11994830 DOI: 10.14309/ajg.0000000000003132] [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: 06/17/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Quality of life (QOL) is critical in screening and management of chronic medical conditions, including celiac disease (CD). The aim of this project was to develop a CD-specific pediatric QOL measure (Celiac Disease Life Inventory of Family Experiences [CDLIFE]) with parallel self-report and parent-report forms by generating items through concept elicitation interviews, iterative refinement using cognitive debriefing interviews, and evaluating its psychometric properties and validity. METHODS Concept elicitation interviews were conducted to develop items (9 youth ages 8-19 years with CD; 10 parents of youth with CD), followed by cognitive interviews with additional stakeholders (3 youth with CD, 3 parents, and 8 clinicians) and item administration (parent/youth reports: n = 103/102). Analyses included response frequencies, internal consistency reliability, exploratory factor analyses, and correlations with related measures (Patient-Reported Outcomes Measurement Information System, Pediatric Quality of Life Inventory, and Gluten-Free Diet-Visual Analog Scale). RESULTS From concept elicitation interviews, 42 youth and 45 parent items were developed. Cognitive debriefing interviews yielded 36 refined items. Psychometric analyses identified 15 items to remove due to ceiling/floor effects, poor item-to-test correlations, and youth-parent mismatch or conceptual mismatch. Total score internal consistency was high (αs = 0.89-0.90). A 4-factor model solution had the best fit (Social Impact, External Support, Adaptive Vigilance, Eating Behaviors/Adjustment) with a fifth single-item domain (Financial Resources). The final CDLIFE (21 items) total scores correlated with most related measures in expected directions for parent and youth forms. DISCUSSION The CDLIFE may facilitate insight into CD-specific QOL for youth ages 2-18 years, capturing important dimensions of physical and socioemotional health. Administering the CDLIFE will help identify and track families needing support.
Collapse
Affiliation(s)
- Shayna Coburn
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
- The George Washington School of Medicine and Health Sciences, 2300 I St NW, Washington, DC 20052
| | - Paige J. Trojanowski
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
| | - Jack Vagadori
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
| | - Pamela Hinds
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
- The George Washington School of Medicine and Health Sciences, 2300 I St NW, Washington, DC 20052
- Department of Nursing Science, Professional Practice & Quality, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
| | - Monique Germone
- Digestive Health Institute, Children’s Hospital Colorado, 13123 E. 16 Avenue, Aurora, CO 80045/ University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO 80045
| | - Edwin Liu
- Digestive Health Institute, Children’s Hospital Colorado, 13123 E. 16 Avenue, Aurora, CO 80045/ University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO 80045
| | - Randi Streisand
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
- The George Washington School of Medicine and Health Sciences, 2300 I St NW, Washington, DC 20052
| | - James Bost
- Center for Translational Research, Children’s National Hospital, 111 Michigan Ave NW, Washington, D.C., 20010
- The George Washington School of Medicine and Health Sciences, 2300 I St NW, Washington, DC 20052
| |
Collapse
|
2
|
Schirru E, Rossino R, Jores RD, Corpino M, Muntoni S, Cucca F, Congia M. Clinical settings in which human leukocyte antigen typing is still useful in the diagnosis of celiac disease. World J Gastroenterol 2025; 31:104397. [PMID: 40248378 PMCID: PMC12001201 DOI: 10.3748/wjg.v31.i14.104397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/01/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Celiac disease (CD) is a systemic autoimmune disorder triggered by gluten ingestion ingenetically predisposed individuals. It is characterized by intestinal histological damage and the production of specific autoantibodies. The latest European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) 2020 guidelines have excluded human leukocyte antigen (HLA) genotyping from the no-biopsy diagnostic approach due to its weak positive predictive value, limited availability, and high cost in some countries. However, HLA genetic testing remains valuable in certain clinical contexts. This study provided practical indications for when to request and how to interpret HLA genotyping, emphasizing its continued relevance for CD diagnosis in specific cases. We also proposed a strategy for monitoring the risk of developing type 1 diabetes (T1D) in patients with CD, based on the risk stratification carried by different HLA genotypes. A retrospective analysis of 746 patients with CD and 627 controls was conducted at our hospital starting in 2012, when HLA genotyping became mandatory for the diagnosis of CD. We identified key clinical scenarios where HLA testing remains useful. Several high risk HLA-DQ genotypes strongly associated with CD were highlighted, including HLA-DQ2.5/HLA-DQ2.2 and HLA-DQ2.5/HLA-DQ2.5. Notably, while the HLA-DQ2.5/HLA-DQ2.2 genotype is linked to CD, it appears to confer protection against T1D. To support clinical practice, we presented a table clarifying commonly used HLA terminology, and another summarized the main clinical situations in which HLA genotyping should still be considered. These findings underscore the dual role of HLA testing: Not only can it help rule out CD in selected cases, but it also identifies patients with CD at risk for T1D, guiding personalized monitoring strategies.
Collapse
Affiliation(s)
- Enrico Schirru
- University Service Center for Animal Facility (CeSASt), University of Cagliari, Monserrato 09042, Sardinia, Italy
| | - Rossano Rossino
- Department of Medical Science and Public Health, University of Cagliari, Monserrato 09042, Sardegna, Italy
- Department of Pediatrics, Clinic of Pediatric and Rare Diseases, Microcitemico Pediatric Hospital, A.Cao, ASL8, Cagliari 09121, Sardegna, Italy
| | - Rita D Jores
- Department Outpatient Clinic, ASL8 Outpatient Clinic, Quartu Sant’Elena 09045, Sardegna, Italy
| | - Mara Corpino
- Department of Pediatrics, Clinic of Pediatric and Rare Diseases, Microcitemico Pediatric Hospital, A.Cao, ASL8, Cagliari 09121, Sardegna, Italy
| | - Sandro Muntoni
- Department of Biomedical Science, University of Cagliari, Monserrato 09042, Sardegna, Italy
| | - Francesco Cucca
- Department of Biomedical Science, University of Sassari, Sassari 07100, Sardegna, Italy
| | - Mauro Congia
- Department of Pediatrics, Clinic of Pediatric and Rare Diseases, Microcitemico Pediatric Hospital, A.Cao, ASL8, Cagliari 09121, Sardegna, Italy
| |
Collapse
|
3
|
Naredi Scherman M, Melin J, Agardh D. Celiac disease screening in children: evaluating the evidence, benefits, and challenges. Front Pediatr 2025; 13:1562073. [PMID: 40248017 PMCID: PMC12003263 DOI: 10.3389/fped.2025.1562073] [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: 01/16/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Comprehensive screening of the general population is the only approach capable of identifying the majority of cases with celiac disease. In 2023, the Italian Parliament enacted a law to implement nationwide screening for celiac disease and type 1 diabetes. However, critical decisions regarding the target population, optimal timing, and screening methods remain unresolved. Previous observational studies on birth cohorts of children with genetic risk for these conditions have demonstrated that the incidence peaks early in life and is influenced by HLA risk genotypes. This mini-review explores different aspects of screening for celiac disease, presenting the advantages and challenges of identifying children before onset of symptoms. In addition, we summarize the current knowledge and gaps in understanding related to screening programs for celiac disease in children and adolescents and discuss health benefits, psychosocial aspects and cost-effectiveness, and their potential implications for future public health strategies.
Collapse
Affiliation(s)
| | | | - Daniel Agardh
- Celiac Disease and Diabetes Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
4
|
Kelley K, Dogru D, Huang Q, Yang Y, Palm NW, Altindis E, Ludvigsson J. Children who develop celiac disease are predicted to exhibit distinct metabolic pathways among their gut microbiota years before diagnosis. Microbiol Spectr 2025; 13:e0146824. [PMID: 39902908 PMCID: PMC11878042 DOI: 10.1128/spectrum.01468-24] [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: 06/18/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Celiac disease (CD) is an autoimmune disease caused by a loss of gluten tolerance in genetically predisposed individuals. While 30%-40% of people possess the predisposing alleles, only 1%-2% are diagnosed with CD, suggesting that environmental factors are involved in disease pathogenesis. To determine an association between pediatric CD and the gut microbiome, we analyzed fecal samples from a prospective cohort study (ABIS). These samples were collected from children who later developed CD (CD progressors) and age-matched healthy children (at ages 1, 2.5, and 5) with similar HLA genotypes, breastfeeding durations, and gluten exposure times. We previously reported gut microbiome differences at ages 2.5 and 5 in this cohort; here, we present findings from samples collected at age 1 (n = 5). We identified 14 ASVs differing significantly between CD progressors and controls, including taxa linked to CD pathogenesis. CD progressors had increased Firmicutes and higher alpha diversity in IgA- bacteria. Using PICRUSt, we analyzed metabolic pathways enriched in CD progressors compared to controls at ages 1, 2.5, and 5 (n = 5-16), revealing enriched inflammatory and pathogenic pathways potentially contributing to CD-related immune dysregulation. While results are based on the primary EdgeR analysis, we also applied a non-parametric method of statistical analysis, reporting those results with supplementary figures. In conclusion, our findings suggest distinct metabolic pathways enriched in the gut microbiome of CD progressors years before diagnosis, which could inform targeted therapeutics for CD. As discussed in the limitations section, this small pilot study should be replicated with larger sample sizes for broader generalization. IMPORTANCE We analyzed gut microbiome data from children who later developed celiac disease (CD progressors) compared to healthy children in the first 5 years of life. Using fecal samples corresponding to the three phases of gut microbiome development, we uncovered enriched functional microbial pathways in CD progressors at age 1. Some of these pathways, implicated in bacterial pathogenesis, microbiota modulation, and inflammation, have been correlated with CD. We also identified taxa in CD progressors at age 1 including Lachnospiraceae, Alistipes, and Bifidobacterium dentium that were previously associated with CD. These findings suggest a potential role for these taxa and enriched pathways in pediatric CD onset years before diagnosis, highlighting potential for early interventions. While the findings of this exploratory study should be validated with larger sample sizes, our study suggests microbial metabolic pathways related to CD onset, enhancing our understanding of CD pathogenesis and the role of gut microbiome-mediated early alterations.
Collapse
Affiliation(s)
- Kristina Kelley
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Dogus Dogru
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Qian Huang
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Johnny Ludvigsson
- Crown Princess Victoria’s Children’s Hospital, Region Östergötland, Linköping, Sweden
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
Corrado MM, Liu E, McQueen RB, Stahl MG. Cost-Minimization Analysis of Celiac Disease Screening Strategies. Pediatrics 2025; 155:e2024068392. [PMID: 39837496 DOI: 10.1542/peds.2024-068392] [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: 07/18/2024] [Accepted: 11/19/2024] [Indexed: 01/23/2025] Open
Affiliation(s)
- Michelle M Corrado
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| | - Edwin Liu
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| | - R Brett McQueen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Marisa G Stahl
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| |
Collapse
|
6
|
Hård Af Segerstad EM, Borge TC, Guo A, Mårild K, Stene LC, Brantsæter AL, Størdal K. Associations of Pregnancy Dietary Quality and Diversity with Childhood Celiac Disease. J Nutr 2024; 154:3770-3779. [PMID: 39428068 DOI: 10.1016/j.tjnut.2024.10.033] [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: 07/01/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND High gluten and low dietary fiber in pregnancy intake is associated with an increased risk of celiac disease (CeD) in the child. Early life higher dietary quality is suggested to reduce the subsequent risk of CeD. OBJECTIVES The aim was to investigate associations of pregnancy dietary quality and diversity with child risk of CeD. METHODS In The Norwegian Mother, Father and Child Cohort Study, 85,122 mother-child pairs had available data from a validated pregnancy food frequency questionnaire. Pregnancy dietary quality and diversity were estimated by a Pregnancy Healthy Eating Index [mean 99.3, standard deviation (SD) 9.9, range 48.8-128.3], and a Diet Diversity Score (mean 7.0, SD 1.0, range 1.6-9.8), respectively. Child CeD was captured by ≥2 diagnostic codes in the Norwegian Patient Registry. Logistic regression was used to estimate associations between pregnancy dietary quality, diversity and child CeD, adjusted for socioeconomic factors, and parents CeD [adjusted odds ratio (aOR), 95% confidence intervals (CI)]. CeD-susceptible human leukocyte antigen haplotypes (DQ2/DQ8) were present in 30,718 (45.5%). RESULTS Up to mean age 16.0 (SD 1.8, 12.4-19.8) y, 1363 (1.6%) children were diagnosed with CeD. Lower as well as higher pregnancy dietary quality associated with a reduced risk of CeD in the child (<5th percentile aOR = 0.67, 95% CI: 0.48, 0.93, >95th percentile aOR = 0.71, 95% CI: 0.52, 0.98, respectively, nonlinear squared term P = 0.011). Analyses on genetically susceptible children, adjustments for pregnancy iron supplementation, gluten, and dietary fiber intake, and child early life dietary quality, gluten intake and iron supplementation, supported the finding. Pregnancy dietary diversity was not associated with child CeD (aOR = 1.00, 95% CI: 0.94, 1.07/score). CONCLUSIONS In this population-based study, lower as well as higher pregnancy dietary quality associated with a reduced risk of CeD diagnosis in the child. In contrast, no such association was observed with maternal dietary diversity.
Collapse
Affiliation(s)
- Elin M Hård Af Segerstad
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway; Unit for Celiac and Diabetes Research, Clinical Sciences, Lund University, Malmoe, Sweden.
| | - Tiril Cecilie Borge
- Cluster for Reviews and Health Technology Assessments, Norwegian Institute of Public Health, Oslo, Norway
| | - Annie Guo
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 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
| | - Lars C Stene
- Cluster for Reviews and Health Technology Assessments, Norwegian Institute of Public Health, Oslo, Norway
| | - Anne Lise Brantsæter
- Department of Food Safety, Center for Sustainable Diets, Norwegian Institute of Public Health, Oslo, Norway
| | - Ketil Størdal
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway; Faculty of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Stanciu D, Staykov H, Dragomanova S, Tancheva L, Pop RS, Ielciu I, Crișan G. Gluten Unraveled: Latest Insights on Terminology, Diagnosis, Pathophysiology, Dietary Strategies, and Intestinal Microbiota Modulations-A Decade in Review. Nutrients 2024; 16:3636. [PMID: 39519469 PMCID: PMC11547711 DOI: 10.3390/nu16213636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A decade of research on gluten-related disorders (GRDs) is reviewed in this study, with a particular emphasis on celiac disease (CD) and non-celiac gluten sensitivity (NCGS). GRDs are triggered by the ingestion of gluten and gluten-like proteins found in wheat, barley, and rye. These proteins lead to intestinal damage in celiac disease, an autoimmune condition characterized by villous atrophy and a variety of gastrointestinal and extraintestinal symptoms. More enigmatic and less understood, NCGS involves symptoms similar to CD but without the immunological reaction or intestinal damage. Recent years have seen advances in the understanding of GRDs, particularly in connection to how intestinal microbiota influences disease progression and patient outcomes. The gluten-free diet (GFD) is still the standard therapy recommended for GRDs despite significant challenges, as discussed in this article. Precise diagnostic methods, patient education and dietary counseling are critical for improving patients' quality of life. The purpose of this review is to provide a more clear and up-to-date understanding of GRDs, and to help further research on this important topic.
Collapse
Affiliation(s)
- Dana Stanciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Hristian Staykov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Radu Samuel Pop
- 3rd Department of Pediatrics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania;
| | - Irina Ielciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Gianina Crișan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| |
Collapse
|
8
|
Punia P, Bala K, Nandi A, Mittal S, Griwan A, Parmar A, Kumar D. Recent Trends in Seroprevalence of Celiac Disease at a Tertiary Care Center. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S2056-S2058. [PMID: 39346472 PMCID: PMC11426858 DOI: 10.4103/jpbs.jpbs_5_24] [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: 01/02/2024] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 10/01/2024] Open
Abstract
It is a retrospective study and all samples received in the laboratory from symptomatic patients for estimation of Immunoglobulin A (IgA) antitissue transglutaminase (IgA anti-tTGA) antibodies are included. Seroprevalence of celiac disease was determined using indirect enzyme-linked immunosorbent assay for IgA anti-tTGA. Out of 8787 serum samples received in the laboratory over a period of four years, the seroprevalence of CD was 2.1, 1.62, 0.72, and 3.3%, respectively.
Collapse
Affiliation(s)
- Parul Punia
- Department of Microbiology, PGIMS, Rohtak, Haryana, India
| | - Kiran Bala
- Department of Microbiology, PGIMS, Rohtak, Haryana, India
| | - Ankita Nandi
- Department of Microbiology, PGIMS, Rohtak, Haryana, India
| | - Seema Mittal
- Department of Microbiology, BPS GMC Khanpurkalan, Sonepat, Haryana, India
| | - Aditya Griwan
- Department of Microbiology, PGIMS, Rohtak, Haryana, India
| | - Aparna Parmar
- Department of Microbiology, PGIMS, Rohtak, Haryana, India
| | - Davender Kumar
- Department of Orthodontics, PGIDS, Rohtak, Haryana, India
| |
Collapse
|
9
|
Boström M, Brundin C, Björck S, Agardh D. Longitudinal screening of HLA-risk and HLA-nonrisk children for celiac disease to age 15 years: CiPiS study. J Pediatr Gastroenterol Nutr 2024; 78:1143-1148. [PMID: 38477348 DOI: 10.1002/jpn3.12181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVES Autoantibodies against tissue transglutaminase (tTG) are serological markers of celiac disease. The aim was to study the applicability of human leukocyte antigen (HLA)-genotyping and tTG autoantibodies in the screening of celiac disease in a longitudinal birth cohort followed to age 15 years. METHODS Included were 13,860 HLA-DQ-genotyped children at birth and previously invited to a screening at age 3 and 9 years, respectively. HLA-DQB1*02 and/or DQB1*03:02 (HLA-risk) children were compared with non-HLA-DQB1*02 and non-DQB1*03:02 (HLA-nonrisk) children. The present study reinvited 12,948/13,860 (93.4%) children at age 15 years of whom 1056/2374 (44.5%) participated in screening at both age 3 and 9 years. Both immunoglobulin A (IgA) and G (IgG) autoantibodies against tTG were analyzed separately in radiobinding assays. Persistently tTG autoantibody-positive children were examined with intestinal biopsy to confirm the diagnosis of celiac disease. RESULTS At age 3 years, celiac disease was diagnosed in 56/1635 (3.4%) HLA-risk children compared with 0/1824 HLA-nonrisk children (p < 0.001). By age 9 years, celiac disease was diagnosed in 72/1910 (3.8%) HLA-risk children compared with 0/2167 HLA-nonrisk children (p < 0.001). Screening at age 15 years detected 14/1071 (1.3%) HLA-risk children positive for IgA-tTG and/or IgG-tTG of whom 12/1071 (1.1%) remained persistently positive. Among those, 10/1071 (0.9%, 95% confidence interval: 0.4%-1.7%) HLA-risk children were diagnosed with celiac disease compared with 0/1303 HLA-nonrisk children (p < 0.001) and 5/491 (1.0%) were negative in screenings at both 3 and 9 years of age. CONCLUSIONS Screening for celiac disease needs to be performed at multiple timepoints to detect all cases but can be restricted to children at HLA-risk.
Collapse
Affiliation(s)
- Michaela Boström
- Celiac Disease and Diabetes Unit, Lund University, Malmö, Sweden
| | | | - Sara Björck
- Celiac Disease and Diabetes Unit, Lund University, Malmö, Sweden
| | - Daniel Agardh
- Celiac Disease and Diabetes Unit, Lund University, Malmö, Sweden
| |
Collapse
|
10
|
Stahl M, Koletzko S, Andrén Aronsson C, Lindfors K, Liu E, Agardh D. Coeliac disease: what can we learn from prospective studies about disease risk? THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:63-74. [PMID: 37972632 PMCID: PMC10965251 DOI: 10.1016/s2352-4642(23)00232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 11/19/2023]
Abstract
Paediatric prospective studies of coeliac disease with longitudinal collection of biological samples and clinical data offer a unique perspective on disease risk. This Review highlights the information now available from international paediatric prospective studies on genetic and environmental risk factors for coeliac disease. In addition, recent omics studies have made it possible to study complex interactions between genetic and environmental factors and thereby further our insight into the causes of the disease. In the future, paediatric prospective studies will be able to provide more detailed risk prediction models combining genes, the environment, and biological corroboration from multiomics. Such studies could also contribute to biomarker development and an improved understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Marisa Stahl
- Pediatric Gastroenterology, Hepatology, and Nutrition, Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Sibylle Koletzko
- Department of Pediatrics, Dr von Hauner Kinderspital, LMU University Hospital, LMU Munich, Munich, Germany; Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Carin Andrén Aronsson
- Unit of Celiac Disease and Diabetes, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Edwin Liu
- Pediatric Gastroenterology, Hepatology, and Nutrition, Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel Agardh
- Unit of Celiac Disease and Diabetes, Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
11
|
Suroviaková S, Ďurdíková A, Ďurdík P, Havličeková Z, Michnová Z, Šutvajová D, Remeň L, Kolková Z, Vojtková J, Bánovčin P. Polysomnographic profile in children diagnosed with celiac disease before starting on a gluten free diet. Sleep Med 2023; 112:301-307. [PMID: 37952478 DOI: 10.1016/j.sleep.2023.10.007] [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: 07/15/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 11/14/2023]
Abstract
STUDY AIMS The study assessed the presence of sleep abnormalities in children who had recently been diagnosed with celiac disease (CD) and not started a gluten free diet (GFD). The children's polysomnographic profiles were also characterized and further compared with healthy children of the same age. METHODS This prospective cross-sectional study involved 46 pediatric subjects (aged 1-19 years) who had recently been diagnosed with CD and not started a GFD. The control group consisted of 32 healthy children (aged 2-17 years). All children underwent anthropometric measurement, laboratory testing and standard overnight observation with in-laboratory video-PSG. The study and control group were divided into subgroups according to the subjects' median ages (8.1 years): celiac children aged less than 8.1 years (n = 23) and more than 8.1 years (n = 23), healthy children less aged than 8.1 years (n = 16) and more than 8.1 years (n = 16). RESULTS No significant differences in the basic demographic and anthropometric parameters between the celiac and control group were observed. Significantly prolonged sleep latency (SOL) was evident in the celiac subjects (21.89 ± 20.77 min. vs. 10.99 ± 7.94 min, p = 0.02), with a probability of prolonged SOL of 4.23-fold greater (OR = 4.23; 95 % CI 1.1-16.22) than the healthy controls, especially in the subgroup of older celiac patients. No significant differences in the sleep period time (SPT), total sleep time (TST), wake during sleep (WASO), sleep efficiency (SE) and sleep stage distribution and cyclization were found. The respiratory rates during sleep indicated a significantly greater incidence of the central apnea-hypopnea index (CAHI) (0.54 ± 0.78 vs. 0.18 ± 0.24, p = 0.03) with a 3.16-fold greater probability of pathological CAHI (OR = 3.16; 95 % CI 1.02-9.77) than the control group. An increased incidence of CSA in the subgroup of younger celiac patients compared to younger healthy controls was especially evident. CONCLUSIONS The findings of our study suggest a difference in sleep architecture and an increased incidence of CSA in children with untreated CD, but additional research is required to verify the results.
Collapse
Affiliation(s)
- Stanislava Suroviaková
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Anna Ďurdíková
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia.
| | - Peter Ďurdík
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Zuzana Havličeková
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Zuzana Michnová
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Dominika Šutvajová
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Lukáš Remeň
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Zuzana Kolková
- Division of Oncology, Biomedical Centre Martin JFM CU, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, 036 01, Martin, Slovakia
| | - Jarmila Vojtková
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| | - Peter Bánovčin
- Department of Pediatrics, Comenius University Bratislava, Jessenius Faculty of Medicine and University Hospital, 036 01, Martin, Slovakia
| |
Collapse
|
12
|
Vauquelin B, Rivière P. [Celiac disease]. Rev Med Interne 2023; 44:539-545. [PMID: 37558601 DOI: 10.1016/j.revmed.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023]
Abstract
Celiac disease is a frequent auto-immune disease characterized by villous atrophy related to gluten intake in patients with genetic susceptiblity. Patients do not present symptoms in the majority of cases. Presence of the disease must be investigated in case of digestive symptoms or presence of auto-immune disease. Diagnosis is based on anti-transglutaminase antibody and dudodenal biospies. The only available treatment is gluten-free diet. Associated auto-immune diseases must be investigated, especially thyroiditis. Complications related to nutritional deficiency must be accounted for also.
Collapse
Affiliation(s)
- B Vauquelin
- Service d'hépato-gastroentérologie et oncologie digestive, centre médico-chirurgical Magellan, CHU de Bordeaux, Bordeaux, France
| | - P Rivière
- Service d'hépato-gastroentérologie et oncologie digestive, centre médico-chirurgical Magellan, CHU de Bordeaux, Bordeaux, France.
| |
Collapse
|
13
|
Urbonas V, Sadauskaite J, Varnas D. Population-Based Screening for Coeliac Disease in Lithuanian Children from 2009 to 2014. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1630. [PMID: 37763749 PMCID: PMC10534554 DOI: 10.3390/medicina59091630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives. Coeliac disease is an autoimmune disorder provoked by a dietary group of proteins called gluten in genetically predisposed individuals. Over the past several decades, the prevalence of coeliac disease has been steadily growing and it is now recognized to be occurring worldwide. The prevalence varies greatly between ethnic, racial groups and regionally. Such variability makes local epidemiological studies important for spreading awareness and setting a threshold for suspicion of coeliac disease. We explored the potential application of a quick point-of-care test for the purpose of detecting a presence of IgA class TG2 antibodies for coeliac disease and screening in a Lithuanian pediatric population. Previously, there were no data regarding coeliac disease prevalence in Lithuania. Materials and Methods. Overall, we included 1458 children 11-13 years of age from several Lithuanian schools selected randomly in this study. Utilizing one point-of-care test using a single blood sample taken from a fingertip, we identified the existence of IgA class TG2 antibodies. Only children whose parents gave consent were enrolled in the study. Those with positive IgA class TG2-ab were directed to a tertiary hospital for additional clinical assessment and confirmation of suspected coeliac disease. Results. A total of two (0.14%) of the 1458 enrolled children were detected with the presence of TG2 antibodies and the coeliac disease diagnosis was further confirmed with histological examination of duodenal biopsy samples. Additionally, we checked that patients had not previously reported any clinical symptoms and signs that could suggest coeliac disease or any other disease of the gastrointestinal tract. Conclusions. The detected prevalence of coeliac disease in the Lithuanian pediatric population is 1:729. The rapid finger prick test for the presence of IgA class TG2 antibodies is a reasonable and accurate method to screen for celiac disease in children.
Collapse
Affiliation(s)
- Vaidotas Urbonas
- Clinic of Children's Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Jolita Sadauskaite
- Clinic of Children's Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Dominykas Varnas
- Clinic of Children's Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| |
Collapse
|
14
|
Martire FG, Piccione E, Exacoustos C, Zupi E. Endometriosis and Adolescence: The Impact of Dysmenorrhea. J Clin Med 2023; 12:5624. [PMID: 37685691 PMCID: PMC10488856 DOI: 10.3390/jcm12175624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Endometriosis affects approximately 10% of premenopausal women worldwide. Despite its impact on quality of life, the delay in diagnosing this chronic disease is well known. Many patients with endometriosis report having suffered from dysmenorrhea and chronic pelvic pain in adolescence or at a young age. However, this painful symptom is often highly underestimated and considered a normal and transient symptom in young women. The real prevalence of endometriosis in adolescence remains uncertain. Some authors recently described at least one ultrasound feature of endometriosis in 13.3% of a general population of adolescent girls, which increased to 35.3% in young girls with severe dysmenorrhea. Dysmenorrhea is classified as primary dysmenorrhea or secondary dysmenorrhea. Primary dysmenorrhea is defined as a menstrual pain without organic disease, while secondary dysmenorrhea is defined as a menstrual pain associated with organic pelvic pathology. Since endometriosis represents the main cause of secondary dysmenorrhea in adolescents and young women, it is important to determine whether the patient has primary dysmenorrhea or additional suggestive symptoms related to endometriosis. Endometriosis in adolescent patients is a challenging problem with clinical and pathological differences compared with its presentation in premenopausal women. Adolescents and young women with dysmenorrhea and painful symptoms that suggest endometriosis should be referred to dedicated endometriosis centers for an early diagnosis and appropriate medical and surgical management. This paper aims to describe the role of dysmenorrhea in adolescents and the management of these young patients to confirm or exclude endometriosis.
Collapse
Affiliation(s)
- Francesco G. Martire
- Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, 53100 Siena, Italy;
| | - Emilio Piccione
- Department of Surgical Sciences, Catholic University “Our Lady of Good Counsel”, 1000 Tirane, Albania
| | - Caterina Exacoustos
- Department of Surgical Sciences, Section of Gynecology and Obstetrics, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Errico Zupi
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
15
|
Lazzeri L, Andersson KL, Angioni S, Arena A, Arena S, Bartiromo L, Berlanda N, Bonin C, Candiani M, Centini G, Forno SD, Donati A, Exacoustos C, Fuggetta E, Labanca L, Maiorana A, Maneschi F, Mattei A, Muzii L, Ottolina J, Perandini A, Perelli F, Pino I, Porpora MG, Remorgida V, Scaramuzzino S, Schimberni M, Seracchioli R, Solima E, Vignali M, Zupi E, Martire FG. How to Manage Endometriosis in Adolescence: The Endometriosis Treatment Italian Club Approach. J Minim Invasive Gynecol 2023; 30:616-626. [PMID: 37001691 DOI: 10.1016/j.jmig.2023.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
The evaluation of endometriosis in an adolescent girl is a challenging topic. The initial stage of the disease and the limited diagnostic instrument appropriate for the youth age and for its typical features can reduce the ability of the gynecologist. At the same time, missing a prompt diagnosis can delay the beginning of specific and punctual management of endometriosis, which could avoid a postponed diagnosis from 6 to 12 years, typical of adolescent girls complaining of dysmenorrhea. This article aimed to answer all the potential questions around the diagnosis and management of endometriosis in adolescents starting from a clinical case looking at the possible solution that is easily reproducible in the clinical practice.
Collapse
Affiliation(s)
- Lucia Lazzeri
- Department of Molecular and Developmental Medicine (Drs. Lazzeri, Centini, Martire, and Zupi), Università di Siena, Siena, Italy
| | - Karin Louise Andersson
- Department of Territory Health (Dr. Andersson, Exacoustos), Azienda Sanitaria Toscana Centro, Florence, Italy
| | - Stefano Angioni
- Department of Surgical Sciences (Dr. Angioni), Università di Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Alessandro Arena
- Department of Medical and Surgical Sciences (Drs. A. Arena, Del Forno, and Seracchioli), DIMEC, Sant'Orsola Hospital, Università di Bologna, Bologna, Italy
| | - Saverio Arena
- Department of Obstetrics and Gynecology (Arena), Santa Maria della Misericordia hospital, Perugia, Italy
| | - Ludovica Bartiromo
- Department of Obstetrics and Gynecology (Drs. Bartiromo, Candiani, Ottolina, and Schimberni), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Berlanda
- Department of Obstetrics and Gynecology (Drs. Berlanda and Donati), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Cecilia Bonin
- Azienda Ospedaliera Universitaria Integrata (Drs. Bonin and Perandini), Università di Verona, Piazzale A. Stefani 1, Verona, Italy
| | - Massimo Candiani
- Department of Obstetrics and Gynecology (Drs. Bartiromo, Candiani, Ottolina, and Schimberni), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Centini
- Department of Molecular and Developmental Medicine (Drs. Lazzeri, Centini, Martire, and Zupi), Università di Siena, Siena, Italy
| | - Simona Del Forno
- Department of Medical and Surgical Sciences (Drs. A. Arena, Del Forno, and Seracchioli), DIMEC, Sant'Orsola Hospital, Università di Bologna, Bologna, Italy
| | - Agnese Donati
- Department of Obstetrics and Gynecology (Drs. Berlanda and Donati), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Caterina Exacoustos
- Department of Territory Health (Dr. Andersson, Exacoustos), Azienda Sanitaria Toscana Centro, Florence, Italy; Department of Surgical Sciences, Gynecologic Unit (Drs. Exacoustos, and Martire), University of Rome "Tor Vergata" Rome, Italy
| | - Eliana Fuggetta
- Department of Obstetrics and Gynecology (Drs. Fuggetta and Maneschi), San Giovanni Addolorata Hospital (Drs. Labanca and Martire), Roma, Italy
| | - Luca Labanca
- Department of Surgical Sciences (Drs. Labanca), Valdarno Hospital, Azienda Toscana Sud Est, Italy
| | - Antonio Maiorana
- Department of Obstetrics and Gynecology (Dr. Maiorana), ARNAS Ospedale Civico Piazza Nicola, Palermo, Italy
| | - Francesco Maneschi
- Department of Obstetrics and Gynecology (Drs. Fuggetta and Maneschi), San Giovanni Addolorata Hospital (Drs. Labanca and Martire), Roma, Italy
| | - Alberto Mattei
- Department of Molecular and Developmental Medicine (Drs. Lazzeri, Centini, Martire, and Zupi), Università di Siena, Siena, Italy; Department of Surgical Sciences, Gynecologic Unit (Drs. Exacoustos, and Martire), University of Rome "Tor Vergata" Rome, Italy
| | - Ludovico Muzii
- Department of Maternal and Child Health and Urology (Drs. Muzii, Porpora, and Scaramuzzino), Università di Roma La Sapienza, Policlinico Umberto I, Rome, Italy
| | - Jessica Ottolina
- Department of Obstetrics and Gynecology (Drs. Bartiromo, Candiani, Ottolina, and Schimberni), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessio Perandini
- Azienda Ospedaliera Universitaria Integrata (Drs. Bonin and Perandini), Università di Verona, Piazzale A. Stefani 1, Verona, Italy
| | - Federica Perelli
- Division of Gynecology and Obstetrics (Drs. Mattei and Perelli), Santa Maria Annunziata Hospital, USL Toscana Centro, Florence, Italy
| | - Ida Pino
- Preventive Gynecology Unit (Dr. Pino), European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Grazia Porpora
- Department of Maternal and Child Health and Urology (Drs. Muzii, Porpora, and Scaramuzzino), Università di Roma La Sapienza, Policlinico Umberto I, Rome, Italy
| | - Valentino Remorgida
- Unit of Obstetrics and Gynecology (Dr. Remorgida), University of Eastern Piedmont, Novara, Italy
| | - Sara Scaramuzzino
- Department of Maternal and Child Health and Urology (Drs. Muzii, Porpora, and Scaramuzzino), Università di Roma La Sapienza, Policlinico Umberto I, Rome, Italy
| | - Matteo Schimberni
- Department of Obstetrics and Gynecology (Drs. Bartiromo, Candiani, Ottolina, and Schimberni), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Renato Seracchioli
- Department of Medical and Surgical Sciences (Drs. A. Arena, Del Forno, and Seracchioli), DIMEC, Sant'Orsola Hospital, Università di Bologna, Bologna, Italy; Division of Gynecology and Human Reproduction Phisiopatology (Dr. Seracchioli), IRCCS, Azienda Ospedaliera Universitaria di Bologna, Bologna Italy
| | - Eugenio Solima
- Department of Obstetrics and Gynecology (Drs. Solima and Vignali), Macedonio Melloni Hospital, Milan, Italy
| | - Michele Vignali
- Department of Obstetrics and Gynecology (Drs. Solima and Vignali), Macedonio Melloni Hospital, Milan, Italy
| | - Errico Zupi
- Department of Molecular and Developmental Medicine (Drs. Lazzeri, Centini, Martire, and Zupi), Università di Siena, Siena, Italy.
| | - Francesco Giuseppe Martire
- Division of Gynecology and Obstetrics (Drs. Mattei and Perelli), Santa Maria Annunziata Hospital, USL Toscana Centro, Florence, Italy
| |
Collapse
|
16
|
Brady RP, Jensen ET, Rigdon J, Crimmins NA, Mallon D, Dolan LM, Imperatore G, Kahkoska AR, Mottl AK, Honor A, Pettitt DJ, Merjaneh L, Dabelea D, Shah AS. The Frequency of Undiagnosed Celiac Disease in Youth with Type 1 Diabetes and Its Association with Diabetic Retinopathy: The SEARCH for Diabetes in Youth Study. Pediatr Diabetes 2023; 2023:9038795. [PMID: 39845332 PMCID: PMC11753297 DOI: 10.1155/2023/9038795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/07/2023] [Accepted: 05/08/2023] [Indexed: 01/24/2025] Open
Abstract
Aims Celiac disease (CD) in adults with type 1 diabetes has been associated with increased cardiovascular risk and the earlier occurrence of diabetes-associated complications. In the Search for Diabetes in Youth study, we aimed to assess the frequency of CD and the potential for undiagnosed CD among youth with childhood onset type 1 diabetes. In addition, we assessed the burden of cardiovascular risk factors and diabetes-associated complications in youth with type 1 diabetes by CD status and IgA tissue transglutaminase autoantibody (tTGA) levels. Methods 2,444 youths with type 1 diabetes completed a CD questionnaire and underwent tTGA testing. Integrating the celiac disease questionnaire and tTGA results for this cross-sectional analysis, participants were categorized as follows: (1) reported CD; (2) seropositive for CD (no reported CD and seropositive tTGA); and (3) type 1 diabetes only (comparison group: no reported CD and seronegative tTGA). Subanalyses were performed on those with no reported CD and tTGA ≥10x ULN, designated potentially undiagnosed CD. Cardiovascular risk factors and diabetes-associated complications were evaluated by CD status and tTGA levels utilizing a Poisson model to estimate relative risk. Results Reported CD in youths with type 1 diabetes was 7%. Seropositivity for tTGA with no reported CD was present in 4%, and 1.2% had potentially undiagnosed CD. Youths with potentially undiagnosed CD had a 2.69x higher risk of diabetic retinopathy than comparison group. In addition, CD with tTGA <0.05 (controlled CD) was associated with lower HbA1c. Conclusions Undiagnosed CD is likely present in youths with type 1 diabetes and potentially undiagnosed CD is associated with a higher risk of diabetic retinopathy. These findings indicate the importance of routine screening for CD in type 1 diabetes in youths.
Collapse
Affiliation(s)
- Ryan P. Brady
- Department of Pediatrics, Cincinnati Children's Hospital & University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Elizabeth T. Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Joseph Rigdon
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Nancy A. Crimmins
- Department of Pediatrics, Cincinnati Children's Hospital & University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Daniel Mallon
- Department of Pediatrics, Cincinnati Children's Hospital & University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lawrence M. Dolan
- Department of Pediatrics, Cincinnati Children's Hospital & University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Giuseppina Imperatore
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Anna R. Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amy K. Mottl
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ann Honor
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | | | - Lina Merjaneh
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Dana Dabelea
- Lifeourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Departments of Epidemiology and Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amy S. Shah
- Department of Pediatrics, Cincinnati Children's Hospital & University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
17
|
Stahl M, Li Q, Lynch K, Koletzko S, Mehta P, Gragert L, Norris JM, Andrén Aronsson C, Lindfors K, Kurppa K, Ilonen J, Krischer J, Alkolkar B, Ziegler AG, Toppari J, Rewers M, Agardh D, Hagopian W, Liu E. Incidence of Pediatric Celiac Disease Varies by Region. Am J Gastroenterol 2023; 118:539-545. [PMID: 36219178 PMCID: PMC9991947 DOI: 10.14309/ajg.0000000000002056] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The Environmental Determinants of Diabetes in the Young study follows an HLA risk selected birth cohort for celiac disease (CD) development using a uniform protocol. Children under investigation come from 6 different regions within Europe and the United States. Our aim was to identify regional differences in CD autoimmunity and CD cumulative incidence for children born between 2004 and 2010. METHODS Children (n = 6,628) with DQ2.5 and/or DQ8.1 were enrolled prospectively from birth in Georgia, Washington, Colorado, Finland, Germany, and Sweden. Children underwent periodic study screening for tissue transglutaminase antibodies and then CD evaluation per clinical care. Population-specific estimates were calculated by weighting the study-specific cumulative incidence with the population-specific haplogenotype frequencies obtained from large stem cell registries from each site. RESULTS Individual haplogenotype risks for CD autoimmunity and CD varied by region and affected the cumulative incidence within that region. The CD incidence by age 10 years was highest in Swedish children at 3%. Within the United States, the incidence by age 10 years in Colorado was 2.4%. In the model adjusted for HLA, sex, and family history, Colorado children had a 2.5-fold higher risk of CD compared to Washington. Likewise, Swedish children had a 1.4-fold and 1.8-fold higher risk of CD compared with those in Finland and Germany, respectively. DISCUSSION There is high regional variability in cumulative incidence of CD, which suggests differential environmental, genetic, and epigenetic influences even within the United States. The overall high incidence warrants a low threshold for screening and further research on region-specific CD triggers.
Collapse
Affiliation(s)
- Marisa Stahl
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Qian Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Kristian Lynch
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sibylle Koletzko
- Department of Pediatrics, Dr von Hauner Kinderspital, LMU Klinikum, Munich, Germany
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Pooja Mehta
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Loren Gragert
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | - Katri Lindfors
- Celiac Disease Research Center, Tampere University and Tampere University Hospital
| | - Kalle Kurppa
- Celiac Disease Research Center, Tampere University and Tampere University Hospital
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital
- University of Consortium of Seinäjoki
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Beena Alkolkar
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - Anette-G Ziegler
- Forschergruppe Diabetes e.V. and Institute of Diabetes Research, Helmholtz Zentrum, Munich, Germany
| | - Jorma Toppari
- Institute of Biomedicine, Centre for Integrative Physiology and Pharmacology, Univeristy of Turku, Turku, Finland
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel Agardh
- Diabetes and Celiac Disease, Lund University, Malmo, Sweden
| | - William Hagopian
- Department of Diabetes, Pacific Northwest Research Institute, Seattle, WA, United States
| | - Edwin Liu
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | |
Collapse
|
18
|
Girdhar K, Dogru YD, Huang Q, Yang Y, Tolstikov V, Raisingani A, Chrudinova M, Oh J, Kelley K, Ludvigsson JF, Kiebish MA, Palm NW, Ludvigsson J, Altindis E. Dynamics of the gut microbiome, IgA response, and plasma metabolome in the development of pediatric celiac disease. MICROBIOME 2023; 11:9. [PMID: 36639805 PMCID: PMC9840338 DOI: 10.1186/s40168-022-01429-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Celiac disease (CD) is an autoimmune disorder triggered by gluten consumption. Almost all CD patients possess human leukocyte antigen (HLA) DQ2/DQ8 haplotypes; however, only a small subset of individuals carrying these alleles develop CD, indicating the role of environmental factors in CD pathogenesis. The main objective of this study was to determine the contributory role of gut microbiota and microbial metabolites in CD onset. To this end, we obtained fecal samples from a prospective cohort study (ABIS) at ages 2.5 and 5 years. Samples were collected from children who developed CD after the final sample collection (CD progressors) and healthy children matched by age, HLA genotype, breastfeeding duration, and gluten-exposure time (n=15-16). We first used 16S sequencing and immunoglobulin-A sequencing (IgA-seq) using fecal samples obtained from the same children (i) 16 controls and 15 CD progressors at age 2.5 and (ii) 13 controls and 9 CD progressors at age 5. We completed the cytokine profiling, and plasma metabolomics using plasma samples obtained at age 5 (n=7-9). We also determined the effects of one microbiota-derived metabolite, taurodeoxycholic acid (TDCA), on the small intestines and immune cell composition in vivo. RESULTS CD progressors have a distinct gut microbiota composition, an increased IgA response, and unique IgA targets compared to healthy subjects. Notably, 26 plasma metabolites, five cytokines, and one chemokine were significantly altered in CD progressors at age 5. Among 26 metabolites, we identified a 2-fold increase in TDCA. TDCA treatment alone caused villous atrophy, increased CD4+ T cells, Natural Killer cells, and two important immunoregulatory proteins, Qa-1 and NKG2D expression on T cells while decreasing T-regulatory cells in intraepithelial lymphocytes (IELs) in C57BL/6J mice. CONCLUSIONS Pediatric CD progressors have a distinct gut microbiota composition, plasma metabolome, and cytokine profile before diagnosis. Furthermore, CD progressors have more IgA-coated bacteria and unique targets of IgA in their gut microbiota. TDCA feeding alone stimulates an inflammatory immune response in the small intestines of C57BJ/6 mice and causes villous atrophy, the hallmark of CD. Thus, a microbiota-derived metabolite, TDCA, enriched in CD progressors' plasma, has the potential to drive inflammation in the small intestines and enhance CD pathogenesis. Video Abstract.
Collapse
Affiliation(s)
- Khyati Girdhar
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | | | - Qian Huang
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | | | - Amol Raisingani
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | | | - Jaewon Oh
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Kristina Kelley
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
| | | | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, 58185, Linköping, SE, Sweden
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
19
|
Andrén Aronsson C, Agardh D. Intervention strategies in early childhood to prevent celiac disease-a mini-review. Front Immunol 2023; 14:1106564. [PMID: 36911718 PMCID: PMC9992640 DOI: 10.3389/fimmu.2023.1106564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
A higher intake of gluten during childhood is associated with increased risk of celiac disease, and the incidence of celiac disease peaks shortly after the time point when associations with higher gluten intake during the second and third year of life occur. Additional environmental factors are most likely necessary for celiac disease to develop. It is hypothesized that gastrointestinal infections increase gut permeability and exposure to gluten. Alternatively, infections may lead to gut dysbiosis and chronic inflammation, with leakage of self-antigens that mimic gluten peptides that leads to an autoimmune-like response. Different gluten interventions to prevent celiac disease have been proposed. Early clinical studies suggested an optimal time point introducing gluten between 4 and 6 months of age while the infant is being breastfed. However, later clinical trials on reduced gluten intake given to infants have shown no protection from celiac disease if gluten introduction was delayed or if gluten was introduced in small amounts during the child's first year of life. Still, more randomized clinical trials (RCTs) are warranted to answer the question if a reduced amount of gluten, not only at the time of introduction during infancy but also in a longer time frame, will prevent children at genetic risk from having lifelong celiac disease. It needs to be clarified whether dietary interventions are effective strategies to be proposed as future prevention of celiac disease in the general population. The present mini-review provides an overview of ongoing or completed RCTs that have focused on interventions during early childhood with the aim of preventing celiac disease.
Collapse
Affiliation(s)
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
20
|
Infantino C, Francavilla R, Vella A, Cenni S, Principi N, Strisciuglio C, Esposito S. Role of Vitamin D in Celiac Disease and Inflammatory Bowel Diseases. Nutrients 2022; 14:nu14235154. [PMID: 36501183 PMCID: PMC9735899 DOI: 10.3390/nu14235154] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin D (VD) is a pro-hormone that has long been known as a key regulator of calcium homeostasis and bone health in both children and adults. In recent years, studies have shown that VD may exert many extra-skeletal functions, mainly through a relevant modulation of the innate and adaptive immune system. This has suggested that VD could play a fundamental role in conditioning development, clinical course, and treatment of several autoimmune disorders, including celiac disease (CD) and inflammatory bowel diseases (IBDs). The main aim of this review is to evaluate the relationships between VD, CD, and IBDs. Literature analysis showed a potential impact of VD on CD and IBDs can be reasonably assumed based on the well-documented in vitro and in vivo VD activities on the gastrointestinal tract and the immune system. The evidence that VD can preserve intestinal mucosa from chemical and immunological damage and that VD modulation of the immune system functions can contrast the mechanisms that lead to the intestinal modifications characteristic of gastrointestinal autoimmune diseases has suggested that VD could play a role in controlling both the development and the course of CD and IBDs. Administration of VD in already diagnosed CD and IBD cases has not always significantly modified disease course. However, despite these relevant problems, most of the experts recommend monitoring of VD levels in patients with CD and IBDs and administration of supplements in patients with hypovitaminosis.
Collapse
Affiliation(s)
- Claudia Infantino
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Roberta Francavilla
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Adriana Vella
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Sabrina Cenni
- Department of Woman, Child and General and Specialized Surgery, Second University of Naples, 80138 Naples, Italy
| | | | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialized Surgery, Second University of Naples, 80138 Naples, Italy
| | - Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
- Correspondence: ; Tel.: +39-0521-704-790
| |
Collapse
|
21
|
Sahin Y, Sevinc E, Bayrak NA, Varol FI, Akbulut UE, Bükülmez A. Knowledge regarding celiac disease among healthcare professionals, patients and their caregivers in Turkey. World J Gastrointest Pathophysiol 2022; 13:178-185. [PMID: 36532302 PMCID: PMC9752282 DOI: 10.4291/wjgp.v13.i6.178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Celiac disease (CD) is one of the most prevalent chronic disorders. The clinical manifestations of CD are diverse and may present with gastrointestinal findings, extra-intestinal findings or no symptoms. Although there has been a marked increase in the prevalence of CD in the past 30 years, up to 95% of patients with CD remain undiagnosed. As most cases have atypical signs or no symptoms, the diagnosis of CD is either missed or delayed. In addition, one of the most important reasons for the delay in diagnosis may be the poor knowledge of healthcare professionals (HCPs) regarding CD. AIM To evaluate the knowledge of HCPs, patients and their caregivers (parents) regarding CD. METHODS The current study was carried out between June 2021 and February 2022 prospectively, as part of the Focus IN CD project. Patients with CD and their caregivers participated in the study from 6 different cities in Turkey. General practitioners, pediatricians, pediatricians with other subspecialities and pediatric gastroenterologists from different cities participated in the study. RESULTS The questionnaire was completed by 348 HCPs, 34 patients with CD, and 102 mothers and 34 fathers of patients with CD. Most of the participants were general practitioners (37.07%). There were 89 (25.57%) pediatricians and 72 (20.69%) pediatric gastroenterologists in the study. The highest score in all categories was achieved by pediatric gastroenterologists. There were significant differences between the four groups of HCPs in terms of the subsections of overall mean score, epidemiology and clinical presentation, treatment and follow-up. No significant difference was found between the groups (patients with CD, mothers of patients with CD and fathers of patients with CD) in terms of the questionnaire subsections. CONCLUSION The level of knowledge on CD among HCPs, patients and their caregivers was unsatisfactory. We consider that it is necessary to increase awareness and to develop e-learning activities on CD among HCPs, patients and their caregivers. Consequently, they may benefit from e-learning programs similar to the one created as part of the EU-funded project Focus IN CD (https://www.celiacfacts.eu/focusincd-en).
Collapse
Affiliation(s)
- Yasin Sahin
- Department of Pediatric Gastroenterology, Dr. Ersin Arslan Training and Research Hospital, Gaziantep, Turkey
- Gaziantep Islam Science and Technology University, Faculty of Medicine, Gaziantep 27560, Gaziantep, Turkey
| | - Eylem Sevinc
- Department of Pediatric Gastroenterology, Karabuk University, Faculty of Medicine, Karabuk 78100, Karabuk, Turkey
| | - Nevzat Aykut Bayrak
- Department of Pediatric Gastroenterology, Zeynep Kamil Women and Children's Training and Research Hospital, University of Health Sciences, Istanbul 34668, Istanbul, Turkey
| | - Fatma Ilknur Varol
- Department of Pediatric Gastroenterology, Inonu University, Faculty of Medicine, Malatya 244280, Malatya, Turkey
| | - Ulas Emre Akbulut
- Department of Pediatric Gastroenterology, University of Health Sciences, Antalya Training and Research Hospital, Antalya 07100, Antalya, Turkey
| | - Ayşegül Bükülmez
- Department of Pediatric Gastroenterology, Afyonkarahisar Health Sciences University, Afyonkarahisar 03200, Afyonkarahisar, Turkey
| |
Collapse
|
22
|
Harju S, Saari A, Sund R, Sankilampi U. Epidemiology of Disorders Associated with Short Stature in Childhood: A 20-Year Birth Cohort Study in Finland. Clin Epidemiol 2022; 14:1205-1214. [PMID: 36320440 PMCID: PMC9618248 DOI: 10.2147/clep.s372870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022] Open
Abstract
Background Many primary and secondary disorders disturb growth and cause short stature (height below −2 SDS) in childhood. Growth monitoring programs aim at their early detection but are not evidence-based: epidemiology of childhood growth disorders is poorly characterized, and no consensus exists on priority target conditions. Herein, we describe population-based epidemiological data on several primary and secondary growth disorders associated with short stature in childhood. Materials and Methods This retrospective population-based 20-year birth cohort study examined 1 144 503 children (51% boys) born in Finland between 1998 and 2017, with 16.5 million care notifications including medical diagnoses. The first occurrences of key primary or secondary growth disorders were identified in multiple registers. Median ages at diagnosis (MAD), and age- and sex-specific cumulative incidences (CMI) from birth until 16 years of age were determined. Results Turner syndrome was the most common primary growth disorder (CMI 52 per 100 000 at 16 years, MAD 4.0 years). Most primary growth disorders were diagnosed before the age of 4 years, and thereafter, secondary growth disorders increased in number. MAD of growth hormone deficiency (GHD) was 8.7 (boys) and 7.2 years (girls). At 16 years, the CMI of GHD was higher in boys than in girls (127 versus 93 per 100 000, respectively), whereas the CMI of hypothyroidism was higher in girls (569 versus 306 per 100 000). Celiac disease was the most common secondary growth disorder and more common in girls than in boys (988 versus 546 per 100 000 at 16 years, respectively). Conclusion These population-based epidemiological data indicate that childhood growth monitoring should be age- and sex-specific. In the early childhood, the focus should be on primary growth disorders, and from preschool age also on secondary growth disorders. These results provide evidence for improving growth monitoring programs and diagnostic practices targeting on Turner syndrome, GHD, hypothyroidism, and celiac disease.
Collapse
Affiliation(s)
- Samuli Harju
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland,Department of Paediatrics, Kuopio University Hospital, Kuopio, Finland,Correspondence: Samuli Harju, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, PO Box 1627, Kuopio, 70211, Finland, Email
| | - Antti Saari
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland,Department of Paediatrics, Kuopio University Hospital, Kuopio, Finland
| | - Reijo Sund
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ulla Sankilampi
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland,Department of Paediatrics, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
23
|
Hård af Segerstad EM, Liu X, Uusitalo U, Agardh D, Andrén Aronsson C, Barbour A, Bautista K, Baxter J, Felipe-Morales D, Stahl BIFM, Gesualdo P, Hoffman M, Karban R, Liu E, Munoz A, Norris J, O'Donnell H, Peacock S, Shorrosh H, Steck A, Stern M, Waugh K, Toppari J, Simell OG, Adamsson A, Aaltonen SM, Ahonen S, Åkerlund M, Hakola L, Hekkala A, Holappa H, Hyöty H, Ikonen A, Ilonen J, Jokipuu S, Karlsson L, Kero J, Koskenniemi JJ, Kähönen M, Knip M, Koivikko ML, Kokkonen K, Koskinen M, Koreasalo M, Kurppa K, Kuusela S, Kytölä J, Laiho J, Latva-aho T, Leppänen L, Lindfors K, Lönnrot M, Mäntymäki E, Mattila M, Miettinen M, Multasuo K, Mykkänen T, Niininen T, Niinistö S, Nyblom M, Oikarinen S, Ollikainen P, Othmani Z, Pohjola S, Rautanen J, Riikonen A, Romo M, Simell S, Tossavainen P, Vähä-Mäkilä M, Varjonen E, Veijola R, Viinikangas I, Virtanen SM, She JX, Schatz D, Hopkins D, Steed L, Bryant J, Silvis K, Haller M, Gardiner M, McIndoe R, Sharma A, Anderson SW, Jacobsen L, Marks J, Towe PD, Ziegler AG, Bonifacio E, Gezginci C, Heublein A, Hohoff E, Hummel S, Knopff A, Koch C, Koletzko S, Ramminger C, Roth R, Schmidt J, et alHård af Segerstad EM, Liu X, Uusitalo U, Agardh D, Andrén Aronsson C, Barbour A, Bautista K, Baxter J, Felipe-Morales D, Stahl BIFM, Gesualdo P, Hoffman M, Karban R, Liu E, Munoz A, Norris J, O'Donnell H, Peacock S, Shorrosh H, Steck A, Stern M, Waugh K, Toppari J, Simell OG, Adamsson A, Aaltonen SM, Ahonen S, Åkerlund M, Hakola L, Hekkala A, Holappa H, Hyöty H, Ikonen A, Ilonen J, Jokipuu S, Karlsson L, Kero J, Koskenniemi JJ, Kähönen M, Knip M, Koivikko ML, Kokkonen K, Koskinen M, Koreasalo M, Kurppa K, Kuusela S, Kytölä J, Laiho J, Latva-aho T, Leppänen L, Lindfors K, Lönnrot M, Mäntymäki E, Mattila M, Miettinen M, Multasuo K, Mykkänen T, Niininen T, Niinistö S, Nyblom M, Oikarinen S, Ollikainen P, Othmani Z, Pohjola S, Rautanen J, Riikonen A, Romo M, Simell S, Tossavainen P, Vähä-Mäkilä M, Varjonen E, Veijola R, Viinikangas I, Virtanen SM, She JX, Schatz D, Hopkins D, Steed L, Bryant J, Silvis K, Haller M, Gardiner M, McIndoe R, Sharma A, Anderson SW, Jacobsen L, Marks J, Towe PD, Ziegler AG, Bonifacio E, Gezginci C, Heublein A, Hohoff E, Hummel S, Knopff A, Koch C, Koletzko S, Ramminger C, Roth R, Schmidt J, Scholz M, Stock J, Warncke K, Wendel L, Winkler C, Lernmark Å, Agardh D, Aronsson CA, Bennet R, Cilio C, Dahlberg S, Fält U, Tsubarah MG, Ericson-Hallström E, Fransson L, Gard T, Halilovic E, Holmén G, Hyberg S, Jonsdottir B, Karimi N, Larsson HE, Lindström M, Lundgren M, Maziarz M, Martinez MM, Melin J, Mestan Z, Nilsson C, Nordh Y, Rahmati K, Ramelius A, Salami F, Sjöberg A, Törn C, Ulvenhag U, Wiktorsson T, Wimar Å, Hagopian WA, Killian M, Crouch CC, Skidmore J, Bowen LS, Metcalf M, Meyer A, Meyer J, Mulenga D, Powell N, Radtke J, Roy S, Schmitt D, Tucker P, Becker D, Franciscus M, Smith MDE, Daftary A, Klein MB, Yates C, Krischer JP, Adusumali R, Austin-Gonzalez S, Avendano M, Baethke S, Burkhardt B, Butterworth M, Cadigan N, Clasen J, Counts K, Gandolfo L, Garmeson J, Gowda V, Karges C, Liu S, Liu X, Lynch K, Malloy J, Mramba L, McCarthy C, Moreno J, Parikh HM, Remedios C, Shaffer C, Smith S, Sulman N, Tamura R, Tewey D, Toth M, Uusitalo U, Vehik K, Vijayakandipan P, Wroble M, Yang J, Young K, Abbondondolo M, Ballard L, Brown R, Cuthbertson D, Dankyi S, Eberhard C, Fiske S, Hadley D, Heyman K, Hsiao B, Laras FP, Lee HS, Li Q, Maguire C, McLeod W, Merrell A, Meulemans S, Quigley R, Smith L, Akolkar B, Yu L, Miao D, Gillespie K, Chandler K, Kelland I, Khoud YB, Randell M, Hagopian W, Radtke J, Tucker P, Erlich H, Mack SJ, Fear AL, Ke S, Mulholland N, Briese T, Brusko T, Johnson SB, McKinney E, Pastinen T, Triplett E. Sources of dietary gluten in the first 2 years of life and associations with celiac disease autoimmunity and celiac disease in Swedish genetically predisposed children: The Environmental Determinants of Diabetes in the Young (TEDDY) study. Am J Clin Nutr 2022; 116:394-403. [PMID: 35394004 PMCID: PMC9348971 DOI: 10.1093/ajcn/nqac086] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND High gluten intake is associated with increased risk of celiac disease (CD) in children at genetic risk. OBJECTIVES We aimed to investigate if different dietary gluten sources up to age 2 y confer different risks of celiac disease autoimmunity (CDA) and CD in children at genetic risk. METHODS Three-day food records were collected at ages 6, 9, 12, 18, and 24 mo from 2088 Swedish genetically at-risk children participating in a 15-y follow-up cohort study on type 1 diabetes and CD. Screening for CD was performed with tissue transglutaminase autoantibodies (tTGA). The primary outcome was CDA, defined as persistent tTGA positivity. The secondary outcome was CD, defined as having a biopsy specimen showing Marsh score ≥ 2 or an averaged tTGA level ≥ 100 Units. Cox regression adjusted for total gluten intake estimated HRs with 95% CIs for daily intake of gluten sources. RESULTS During follow-up, 487 (23.3%) children developed CDA and 242 (11.6%) developed CD. Daily intake of ≤158 g porridge at age 9 mo was associated with increased risk of CDA (HR: 1.53; 95% CI: 1.05, 2.23; P = 0.026) compared with no intake. A high daily bread intake (>18.3 g) at age 12 mo was associated with increased risk of both CDA (HR: 1.47; 95% CI: 1.05, 2.05; P = 0.023) and CD (HR: 1.79; 95% CI: 1.10, 2.91; P = 0.019) compared with no intake. At age 18 mo, milk cereal drink was associated with an increased risk of CD (HR: 1.16; 95% CI: 1.00, 1.33; P = 0.047) per 200-g/d increased intake. No association was found for other gluten sources up to age 24 mo and risk of CDA or CD. CONCLUSIONS High daily intakes of bread at age 12 mo and of milk cereal drink during the second year of life are associated with increased risk of both CDA and CD in genetically at-risk children.
Collapse
Affiliation(s)
| | - Xiang Liu
- Health Informatics Institute, Department of Pediatrics, Morsani Collage of Medicine, University of South Florida, Tampa, FL, USA
| | - Ulla Uusitalo
- Health Informatics Institute, Department of Pediatrics, Morsani Collage of Medicine, University of South Florida, Tampa, FL, USA
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Meijer CR, Auricchio R, Putter H, Castillejo G, Crespo P, Gyimesi J, Hartman C, Kolacek S, Koletzko S, Korponay-Szabo I, Ojinaga EM, Polanco I, Ribes-Koninckx C, Shamir R, Szajewska H, Troncone R, Villanacci V, Werkstetter K, Mearin ML. Prediction Models for Celiac Disease Development in Children From High-Risk Families: Data From the PreventCD Cohort. Gastroenterology 2022; 163:426-436. [PMID: 35487291 DOI: 10.1053/j.gastro.2022.04.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Screening for celiac disease (CD) is recommended in children with affected first-degree relatives (FDR). However, the frequency of screening and at what age remain unknown. The aims of this study were to detect variables influencing the risk of CD development and develop and validate clinical prediction models to provide individualized screening advice. METHODS We analyzed prospective data from the 10 years of follow-up of the PreventCD-birth cohort involving 944 genetically predisposed children with CD-FDR. Variables significantly influencing the CD risk were combined to determine a risk score. Landmark analyses were performed at different ages. Prediction models were created using multivariable Cox proportional hazards regression analyses, backward elimination, and Harrell's c-index for discrimination. Validation was done using data from the independent NeoCel cohort. RESULTS In March 2019, the median follow-up was 8.3 years (22 days-12.0 years); 135/944 children developed CD (mean age, 4.3 years [range, 1.1-11.4]). CD developed significantly more often in girls (P = .005) and in Human Leukocyte Antigen (HLA)-DQ2 homozygous individuals (8-year cumulative incidence rate of 35.4% vs maximum of the other HLA-risk groups 18.2% [P < .001]). The effect of homozygosity DR3-DQ2/DR7-DQ2 on CD development was only present in girls (interaction P = .04). The prediction models showed good fit in the validation cohort (Cox regression 0.81 [0.54]). To calculate a personalized risk of CD development and provide screening advice, we designed the Prediction application https://hputter.shinyapps.io/preventcd/. CONCLUSION Children with CD-FDR develop CD early in life, and their risk depends on gender, age and HLA-DQ, which are all factors that are important for sound screening advice. These children should be screened early in life, including HLA-DQ2/8-typing, and if genetically predisposed to CD, they should get further personalized screening advice using our Prediction application. TRIAL REGISTRATION NUMBER ISRCTN74582487 (https://www.isrctn.com/search?q=ISRCTN74582487).
Collapse
Affiliation(s)
- Caroline R Meijer
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Renata Auricchio
- Translational Medical Sciences and European Laboratory for the Investigation of Food-Induced Disease, University of Naples Federico II, Naples, Italy
| | - Hein Putter
- Department of Medical Statistics, Leiden University Medical Center, Leiden, the Netherlands
| | - Gemma Castillejo
- Pediatric Gastroenterology Unit, Hospital Universitario Sant Joan de Reus, Reus, Spain
| | - Paula Crespo
- ADViSE, Department of Health Sciences, European University Miguel de Cervantes, Hospital Recoletas Campo Grande, Valladolid, Spain
| | - Judit Gyimesi
- Coeliac Disease Centre, Heim Pál National Paediatric Institute, Budapest, Hungary
| | - Corina Hartman
- Institute for Gastroenterology, Nutrition and Liver Disease, Schneider Children's Medical Center, Sackler Faculty of Medicine Tel Aviv University, Tel Aviv, Israel
| | - Sanja Kolacek
- Referral Center Pediatric Gastroenterology and Nutrition, Zagreb University, Medical School, Zagreb, Croatia
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany; Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Ilma Korponay-Szabo
- Coeliac Disease Centre, Heim Pál National Paediatric Institute, Budapest, Hungary
| | - Eva Martinez Ojinaga
- Pediatric Gastroenterology and Nutrition, La Paz University Hospital, Madrid, Spain
| | - Isabel Polanco
- Pediatric Gastroenterology and Nutrition, La Paz University Hospital, Madrid, Spain
| | | | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Disease, Schneider Children's Medical Center, Sackler Faculty of Medicine Tel Aviv University, Tel Aviv, Israel
| | - Hania Szajewska
- Pediatrics, Warsaw, Medical University of Warsaw, Warsaw, Poland
| | - Riccardo Troncone
- Translational Medical Sciences and European Laboratory for the Investigation of Food-Induced Disease, University of Naples Federico II, Naples, Italy
| | | | - Katharina Werkstetter
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany
| | - M Luisa Mearin
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
25
|
Lebwohl B, Greco L. Can We Predict the Onset of Celiac Disease? Gastroenterology 2022; 163:368-369. [PMID: 35661721 DOI: 10.1053/j.gastro.2022.05.045] [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: 05/05/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Benjamin Lebwohl
- The Celiac Disease Center, Columbia University, New York, New York.
| | - Luigi Greco
- European Laboratory for Food Induced Disease, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Germone M, Phu T, Slosky C, Pan Z, Jones A, Stahl M, Mehta P, Shull M, Ariefdjohan M, Liu E. Anxiety and Depression in Pediatric Patients with Celiac Disease: A Large Cross-Sectional Study. J Pediatr Gastroenterol Nutr 2022; 75:181-185. [PMID: 35641896 DOI: 10.1097/mpg.0000000000003497] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Mental health is a growing concern in pediatric celiac disease (CD). This study utilized the Revised Children's Anxiety and Depression Scale (RCADS) to investigate anxiety and depression symptom rates. Participants were children ages 8 to 17 years (M = 11.7, SD = 2.7; N = 175) with biopsy-proven CD (Median = 1.1 years post-diagnosis, IQR = 0-4) categorized into groups based on the child's age, caregiver or child respondent, presence or absence of comorbidities, and gluten-free diet duration. Self-reported RCADS scores showed 39% of children having clinically significant concerns for anxiety or depression ( P < 0.0001) but only 7% of caregiver-proxy RCADS scores indicated significant concerns for the child's anxiety and 14% for the child's depression. Rates of child-reported anxiety and depression symptoms were significantly higher for those without medical comorbidities than those with ( P = 0.04). Therefore, screening for mental health concerns, particularly anxiety and depression, should be routinely performed in pediatric patients with CD.
Collapse
Affiliation(s)
- Monique Germone
- From the Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO
- the Pediatric Mental Health Institute, Children's Hospital Colorado, Aurora, CO
- the Colorado Center for Celiac Disease, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO
| | - Tiffany Phu
- the Pediatric Mental Health Institute, Children's Hospital Colorado, Aurora, CO
| | - Camryn Slosky
- From the Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Zhaoxing Pan
- the Biostatistics Core of Children's Hospital Colorado Research Institute
| | - Anna Jones
- the Department of Psychology, St. Jude Children's Research Hospital, Memphis, TN
| | - Marisa Stahl
- the Colorado Center for Celiac Disease, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO
- the Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Pooja Mehta
- the Colorado Center for Celiac Disease, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO
- the Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mary Shull
- the Colorado Center for Celiac Disease, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO
- the Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Merlin Ariefdjohan
- From the Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Edwin Liu
- the Colorado Center for Celiac Disease, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO
- the Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
27
|
de Sousa Franckilin LR, Dos Santos ACPM, Freitas FEDA, Vieira IG, de Freitas Jorge CE, Neri DG, de Abreu MVC, Fonseca JK, Loffi RG, Foureaux G. Gluten: do only celiac patients benefit from its removal from the diet? FOOD REVIEWS INTERNATIONAL 2022. [DOI: 10.1080/87559129.2021.2024566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | - Renato Guimarães Loffi
- Departamento de Ciência, Tecnologia e Inovação, Treini Biotecnologia Ltda, Belo Horizonte, Brazil
| | - Giselle Foureaux
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Nutrição, Angiogold: Medicina Integrativa, Belo Horizonte, Brazil
| |
Collapse
|
28
|
Family ties: the impact of celiac disease on children and caregivers. Qual Life Res 2022; 31:2107-2118. [PMID: 34984588 DOI: 10.1007/s11136-021-03078-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate the impact of celiac disease (CD) and the gluten-free diet (GFD) on the health-related quality of life (HRQoL) in children with CD in the United States using validated measures. We hypothesize that CD negatively impacts the child and caregivers' HRQoL. METHODS Participants included children with a confirmed diagnosis of CD and their caregivers (n = 246) seen in a CD multidisciplinary clinic. Caregivers completed the Pediatric Quality of Life (PedsQL) parent-proxy scale to report on their child's HRQoL and the Family Impact Module (FIM), which assesses the impact of caring for a child with a chronic illness. Their children completed the age-appropriate PedsQL. PedsQL and FIM results were compared to published data for children with gastroenterological conditions and a healthy cohort using non-parametric tests. RESULTS Children with CD reported significantly lower HRQoL than reports from healthy controls across all PedsQL domains (P < 0.001, Cohen d = 0.8), and lower compared to children with other organic gastrointestinal conditions in Social Functioning (P < 0.001, Cohen d = 0.5) and overall Psychosocial Functioning (P < 0.001, Cohen d = 0.3) domains. Results from the caregiver's report on their own HRQoL were significantly worse than that reported by historical controls in the domains of Communication (P < 0.001, Cohen d = 0.3) and Worry (P < 0.001, Cohen d = 0.8), yet similar on all other domains. CONCLUSIONS In our population, CD is associated with low HRQoL scores for both children and their caregivers. Screening children and families for HRQoL can identify patients and families in need of additional support in this higher-risk population.
Collapse
|
29
|
Kurppa K, Agardh D. Pediatric coeliac disease. COELIAC DISEASE AND GLUTEN-RELATED DISORDERS 2022:23-41. [DOI: 10.1016/b978-0-12-821571-5.00002-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
30
|
Autoimmune Diseases of Digestive Organs-A Multidisciplinary Challenge: A Focus on Hepatopancreatobiliary Manifestation. J Clin Med 2021; 10:jcm10245796. [PMID: 34945093 PMCID: PMC8705412 DOI: 10.3390/jcm10245796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022] Open
Abstract
It is well known that some pathological conditions, especially of autoimmune etiology, are associated with the HLA (human leukocyte antigen) phenotype. Among these diseases, we include celiac disease, inflammatory bowel disease, autoimmune enteropathy, autoimmune hepatitis, primary sclerosing cholangitis and primary biliary cholangitis. Immunoglobulin G4-related diseases (IgG4-related diseases) constitute a second group of autoimmune gastrointestinal, hepatobiliary and pancreatic illnesses. IgG4-related diseases are systemic and rare autoimmune illnesses. They often are connected with chronic inflammation and fibrotic reaction that can occur in any organ of the body. The most typical feature of these diseases is a mononuclear infiltrate with IgG4-positive plasma cells and self-sustaining inflammatory response. In this review, we focus especially upon the hepatopancreatobiliary system, autoimmune pancreatitis and IgG4-related sclerosing cholangitis. The cooperation of the gastroenterologist, radiologist, surgeon and histopathologist is crucial for establishing correct diagnoses and appropriate treatment, especially in IgG4 hepatopancreatobiliary diseases.
Collapse
|
31
|
Sahin Y. Celiac disease in children: A review of the literature. World J Clin Pediatr 2021; 10:53-71. [PMID: 34316439 PMCID: PMC8290992 DOI: 10.5409/wjcp.v10.i4.53] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/23/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Celiac disease is an immune-mediated systemic disease triggered by intake of gluten in genetically susceptible individuals. The prevalence of celiac disease in the general population is estimated to be 1% in the world. Its prevalence differs depending on geographical and ethnic variations. The prevalence of celiac disease has increased significantly in the last 30 years due to the increased knowledge and awareness of physicians and the widespread use of highly sensitive and specific diagnostic tests for celiac disease. Despite increased awareness and knowledge about celiac disease, up to 95% of celiac patients still remain undiagnosed. The presentations of celiac disease have significantly changed in the last few decades. Classical symptoms of celiac disease occur in a minority of celiac patients, while older children have either minimal or atypical symptoms. Serologic tests for celiac disease should be done in patients with unexplained chronic or intermittent diarrhea, failure to thrive, weight loss, delayed puberty, short stature, amenorrhea, iron deficiency anemia, nausea, vomiting, chronic abdominal pain, abdominal distension, chronic constipation, recurrent aphthous stomatitis, and abnormal liver enzyme elevation, and in children who belong to specific groups at risk. Early diagnosis of celiac disease is very important to prevent long-term complications. Currently, the only effective treatment is a lifelong gluten-free diet. In this review, we will discuss the epidemiology, clinical findings, diagnostic tests, and treatment of celiac disease in the light of the latest literature.
Collapse
Affiliation(s)
- Yasin Sahin
- Pediatric Gastroenterology-Hepatology and Nutrition, Medical Park Gaziantep Hospital, Gaziantep 27560, Turkey
| |
Collapse
|
32
|
Torun A, Hupalowska A, Trzonkowski P, Kierkus J, Pyrzynska B. Intestinal Microbiota in Common Chronic Inflammatory Disorders Affecting Children. Front Immunol 2021; 12:642166. [PMID: 34163468 PMCID: PMC8215716 DOI: 10.3389/fimmu.2021.642166] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.
Collapse
Affiliation(s)
- Anna Torun
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Anna Hupalowska
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beata Pyrzynska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
33
|
Abstract
INTRODUCTION To test whether parechovirus and anellovirus, frequent enteric viruses, were associated with subsequent celiac disease (CD). We hypothesized that children who later developed CD would have increased frequency of parechovirus infections before transglutaminase 2 (TG2) antibody development. Anellovirus testing was exploratory, as a potential marker of immune status. METHODS Matched case-control design nested within a longitudinal birth cohort (the MIDIA study) of children at genetic risk of CD (carrying the human leukocyte antigen genotype DR4-DQ8/DR3-DQ2, recruited throughout Norway during 2001-2007). We retrospectively tested blood samples taken at age 3, 6, 9, and 12 months, and then annually, to determine when TG2 antibodies developed. Of 220 genetically at-risk children tested, 25 were diagnosed with CD (cases; ESPGHAN 2012 criteria) and matched for follow-up time, birthdate, and county of residence with 2 randomly selected children free from CD (controls) from the cohort. Viruses were quantified in monthly stool samples (collected from 3 through 35 months of age) using real-time polymerase chain reaction methods. RESULTS Parechovirus was detected in 222 of 2,005 stool samples (11.1%) and was more frequent in samples from cases before developing TG2 antibodies (adjusted odds ratio 1.67, 95% confidence interval 1.14-2.45, P = 0.01). The odds ratio was higher when a sample was positive for both parechovirus and enterovirus (adjusted odds ratio 4.73, 95% confidence interval 1.26-17.67, P = 0.02). Anellovirus was detected in 1,540 of 1,829 samples (84.2%), but did not differ significantly between case and control subjects. DISCUSSION Early-life parechovirus infections were associated with development of CD in genetically at-risk children.
Collapse
|
34
|
Sakhuja S, Holtz LR. Progression of pediatric celiac disease from potential celiac disease to celiac disease: a retrospective cohort study. BMC Pediatr 2021; 21:149. [PMID: 33781221 PMCID: PMC8006356 DOI: 10.1186/s12887-021-02625-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
Background A subset of patients with serology suggesting celiac disease have an initially negative biopsy but subsequently develop histopathologic celiac disease. Here we characterize patients with potential celiac disease who progress to celiac disease. Methods We performed a retrospective analysis of children (0–18 years of age) with biopsy-confirmed celiac disease seen at St. Louis Children’s Hospital between 2013 and 2018. Results Three hundred sixteen of 327 (96%) children with biopsy-confirmed celiac disease were diagnosed on initial biopsy. The 11 children with potential celiac disease who progressed to celiac disease had lower anti-tissue transglutaminase (anti-TTG IgA) concentrations (2.4 (1.6–5) X upper limit of normal (ULN) vs. 6.41 (3.4–10.5) X ULN) at time of first biopsy. Their median anti-TTG IgA concentrations rose from 2.4 (1.6–5) X ULN to 3.6 (3.1–9.2) X ULN between biopsies. Conclusions Four percent of biopsy confirmed celiac patients initially had a negative biopsy, but later developed histopathologic celiac disease. This is likely an underestimate as no surveillance algorithm was in place. We recommend repeat assessment in children whose serology suggests celiac disease despite normal small bowel biopsy. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-02625-z.
Collapse
Affiliation(s)
- Shruti Sakhuja
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid, Campus, Box 8208, St. Louis, MO, 63110, USA
| | - Lori R Holtz
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid, Campus, Box 8208, St. Louis, MO, 63110, USA.
| |
Collapse
|
35
|
Keceli Basaran M, Dogan C, Bal M, Geylani Gulec S, Urganci N. Does Having Rotavirus Infection in Early Childhood Increase the Risk of Celiac Disease? J PEDIAT INF DIS-GER 2021. [DOI: 10.1055/s-0041-1726074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Objective With the increasing prevalence of celiac disease (CD) in the population, possible risk factors are under investigation. Environmental and genetic factors that trigger the immune response have been analyzed for many years. This study investigates the presence of CD in children with rotavirus infection. Rotavirus infection is thought to be a risk factor for CD.
Methods Included in the study were 105 of 160 pediatric patients hospitalized due to symptomatic rotavirus infection between 2012 and 2018. These children were screened for CD 45.6 ± 18.2 (14–90) months following the rotavirus infection diagnosed with CD as per ESPGHAN guidelines.
Results A total of 105 pediatric patients who had rotavirus gastroenteritis were included in the study. The age of the children with rotavirus infection was 3.98 ± 1 (2–6) months. In terms of CD, it was 45.6 ± 18.2 months. Around 14 to 90 months later, patients were called for control. CD developed in four (3.8%) of the children with rotavirus, whereas none of the children in the control group developed CD.
Conclusion Rotavirus infection may be a risk factor for CD through immune mechanisms. There are genetic and various environmental factors for the development of CD. Although the CD's occurrence on children who had rotavirus gastroenteritis in our study also supported this situation, there was no statistically significant difference.
Collapse
Affiliation(s)
- Meryem Keceli Basaran
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gaziosmanpaşa Training and Research Hospital, Istanbul, Turkey
| | - Caner Dogan
- Department of Pediatrics, Gaziosmanpaşa and Research Hospital, Istanbul, Turkey
| | - Mahmut Bal
- Department of Pediatrics, Gaziosmanpaşa and Research Hospital, Istanbul, Turkey
| | - Seda Geylani Gulec
- Department of Pediatrics, Gaziosmanpaşa and Research Hospital, Istanbul, Turkey
| | - Nafiye Urganci
- Department of Pediatrics, Division of Pediatric Gastroenterology, Sariyer Etfal Education and Research Hospital, Istanbul, Turkey
| |
Collapse
|
36
|
Bykova SV, Sabelnikova EA, Gudkova RB, Noskova KK, Krums LM, Parfenov AI. [Screening for celiac disease among patients of the gastroenterological profile]. TERAPEVT ARKH 2021; 93:145-149. [PMID: 36286627 DOI: 10.26442/00403660.2021.02.200625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/05/2021] [Indexed: 12/26/2022]
Abstract
AIM To determine the frequency of celiac disease (CD) among gastroenterological patients and criteria for its active detection. MATERIALS AND METHODS 1.358 patients referred for gastroenterologist consultation from 2016 to 2019 was conducted, of which 140 had CD (339 males 24.9%; 1019 females 75.1%). The average age was 40.415.4 (1886 years). All patients were determined anti-TTG IgA, IgG, and analyzed the clinical symptoms and analysis. The results were subjected to statistical processing Statistica 13.3 (StatSoft Inc., USA). RESULTS In patients without CD (1218 people), high level of anti-TTG IgA and IgG was observed in 59 (4.8%), an increase in anti-TTG IgA in 54 (4.4%), and anti-TTG IgG in 38 patients (3.1%). The CD diagnosis confirmed in 51 patients (4.2%). The main symptoms were diarrhea (88%), abdominal pain (60.7%), bloating (73.8%), nausea (40.3%), weight loss (44.3%). Anemia was determined in 31.6%, serum iron 33%, hypoproteinemia 12.6%, hypoalbuminemia 12%, hypokalemia 5.48%, hypocalcemia 21.9%. An increase in the level of AST 14.5%, ALT 14.6%. Comparative analysis showed that in the group with newly detected CD, anemia, malabsorption syndrome, increase AST, ALT were significantly more frequent than in patients with normal antibodies, which confirms the need to detect CD among patients with these laboratory abnormalities. CONCLUSION The incidence of CD among patients with a gastroenterological symptoms was 4.2%. Analysis of clinical and laboratory data has shown that a comprehensive analysis of clinical symptoms and laboratory indicators at the stage of primary treatment will allow timely identification of CD patients and prescribe GFD.
Collapse
Affiliation(s)
- S V Bykova
- Loginov Moscow Clinical Research and Practical
| | | | - R B Gudkova
- Loginov Moscow Clinical Research and Practical
| | - K K Noskova
- Loginov Moscow Clinical Research and Practical
| | - L M Krums
- Loginov Moscow Clinical Research and Practical
| | | |
Collapse
|
37
|
Abstract
BACKGROUND It is not known if genetic background, characteristics at diagnosis, physical and psychological well-being, and adherence to a gluten-free diet are comparable between patients with familial or sporadic celiac disease. These issues were investigated in a follow-up study. METHODS Altogether 1064 patients were analyzed for celiac disease-associated serology, predisposing HLA-DQ, and non-HLA genotypes. Medical data were collected from patient records and supplementary interviews. Current symptoms and quality of life were further evaluated with the Gastrointestinal Symptom Rating Scale (GSRS), the Psychological General Well-Being questionnaire (PGWB), and Short Form 36 (SF-36) questionnaires. RESULTS Familial and sporadic groups differed (P < 0.001) in the reason for diagnosis and clinical presentation at diagnosis, familial patients being more often screen-detected (26% vs. 2%, P < 0.001) and having less often gastrointestinal (49% vs. 69%) and severe symptoms (47% vs. 65%). The groups were comparable in terms of histological damage, frequency of malabsorption, comorbidities, childhood diagnoses, and short-term treatment response. At the time of the study, familial cases reported fewer symptoms (21% vs. 30%, P = 0.004) and lower prevalence of all (78% vs. 86%, P = 0.007), neurological (10% vs. 15%, P = 0.013), and dermatological (9% vs. 17%, P = 0.001) comorbidities. Dietary adherence and GSRS scores were comparable, but familial cases had better quality of life according to PGWB and SF-36. High-risk genotype HLA-DQ2.5/DQ2.5 was more frequent among familial cases, and four non-HLA SNPs were associated with familial celiac disease. CONCLUSIONS Despite the greater proportion of high-risk genotypes, familial cases had milder symptoms at presentation than did sporadic cases. Worse experience of symptoms and poorer quality of life in sporadic disease indicate a need for intensified support.
Collapse
|
38
|
Mass Screening for Celiac Disease: The Autoimmunity Screening for Kids Study. Am J Gastroenterol 2021; 116:180-187. [PMID: 32701732 PMCID: PMC7775339 DOI: 10.14309/ajg.0000000000000751] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The Autoimmunity Screening for Kids (ASK) study is a large scale pediatric screening study in Colorado for celiac disease (CD) and type 1 diabetes. This is a report of the CD outcomes for the first 9,973 children screened through ASK. METHODS ASK screens children aged 1-17 years for CD using 2 highly sensitive assays for tissue transglutaminase autoantibodies (TGA): a radiobinding (RBA) assay for IgA TGA and an electrochemiluminescence (ECL) assay that detects all TGA isotypes. Children who test positive on either assay are asked to return for confirmatory testing. Those with a confirmed RBA TGA level ≥ 0.1 (twice the upper limit of normal) are referred to the Colorado Center for Celiac Disease for further evaluation; all others are referred to primary care. RESULTS Of the initial 9,973 children screened, 242 children were TGA+ by any assay. Of those initially positive, 185 children (76.4%) have completed a confirmation blood draw with 149 children (80.5%) confirming positive by RBA TGA. Confirmed RBA TGA+ was associated with a family history of CD (odds ratio [OR] = 1.83; 95% confidence interval 1.06-3.16), non-Hispanic white ethnicity (OR = 3.34; 2.32-4.79), and female sex (OR = 1.43; 1.03-1.98). Gastrointestinal symptoms of CD, assessed at the initial screening, were reported equally often among the RBA TGA+ vs TGA- children (32.1% vs 30.5%, P = 0.65). DISCUSSION The initial results of this ongoing mass-screening program confirm a high prevalence of undiagnosed CD autoimmunity in a screened US population. Symptoms at initial screening were not associated with TGA status (see Visual abstract, Supplementary Digital Content 5, http://links.lww.com/AJG/B587).
Collapse
|
39
|
Lebwohl B, Rubio-Tapia A. Epidemiology, Presentation, and Diagnosis of Celiac Disease. Gastroenterology 2021; 160:63-75. [PMID: 32950520 DOI: 10.1053/j.gastro.2020.06.098] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/22/2020] [Accepted: 06/06/2020] [Indexed: 12/16/2022]
Abstract
The incidence of celiac disease is increasing, partly because of improved recognition of, and testing for, the disease. The rise in incidence is also due to a real increase of this immune-based disorder, independent of disease detection. The reasons for this true rise in recent decades are unknown but may be related to environmental factors that may promote loss of tolerance to dietary gluten. Strategies to reduce the development of celiac disease have not been proven successful in randomized trials, but the quantity of early-life gluten exposure has been a major focus of prevention efforts. The criteria for the diagnosis of celiac disease are changing, but in adults, diagnosis still depends on the presence of duodenal villous atrophy while the patient is on a gluten-containing diet, along with findings from serology analysis. Although guidelines in the United States continue to mandate a biopsy at all ages, some children receive a diagnosis of celiac disease without a biopsy. If proven accurate and scalable, assays that detect gluten-HLA tetramer complexes might be used in diagnosis to be made in the context of a gluten-free diet without intestinal biopsy.
Collapse
Affiliation(s)
- Benjamin Lebwohl
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York.
| | - Alberto Rubio-Tapia
- Department of Gastroenterology, Hepatology, and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
40
|
Stahl MG, Dong F, Lamb MM, Waugh KC, Taki I, Størdal K, Stene LC, Rewers MJ, Liu E, Norris JM, Mårild K. Childhood growth prior to screen-detected celiac disease: prospective follow-up of an at-risk birth cohort. Scand J Gastroenterol 2020; 55:1284-1290. [PMID: 32941083 PMCID: PMC7646943 DOI: 10.1080/00365521.2020.1821087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To determine the association between childhood growth prior to the development of celiac disease (CD) and CD autoimmunity (CDA) identified by periodic serological screening. STUDY DESIGN The Diabetes Autoimmunity Study in the Young cohort includes 1979 genetically at-risk children from Denver, Colorado, with annual growth measurements from age nine months until ten years. Between 1993 and February 2019, 120 children developed CDA defined by persistent positive tissue transglutaminase autoantibodies (TGA); among these, 71 met our criteria for CD based on histopathological findings or high TGA levels. Age- and sex-specific z-scores of weight, body mass index (BMI), and height prior to seroconversion were derived using US reference charts as standards. Joint modeling of serial growth measurements was used to estimate adjusted hazard ratios (aHRs) accounting for celiac-associated human leukocyte antigens, early-life feeding practices, and socio-demographics. RESULTS In the first 10 years of life, there were no significant associations between the child's current weight, BMI and height and the risk of screening-detected CDA or CD, neither was the weight nor BMI velocity associated with CDA or CD as identified by screening (all aHRs approximated 1). Increased height velocity was associated with later CD, but not CDA, development (aHR per 0.01-z score/year, 1.28; 95% confidence interval [CI] 1.18-1.38 and 1.03; 0.97-1.09, respectively). CONCLUSIONS In the first 10 years of life, from prospectively collected serial growth measurements, we found no evidence of impaired childhood growth before CD and CDA development as identified through early and periodic screening.
Collapse
Affiliation(s)
- Marisa G. Stahl
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Fran Dong
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Molly M. Lamb
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathleen C. Waugh
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iman Taki
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ketil Størdal
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Pediatrics, Østfold Hospital Trust, Grålum, Norway
| | - Lars C. Stene
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Marian J. Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edwin Liu
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karl Mårild
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg, Sweden
- Department of Pediatric Gastroenterology, Queen Silvia Children’s Hospital, Gothenburg, Sweden
| |
Collapse
|
41
|
Spector Cohen I, Day A, Shaoul R. Should the Glu Be Ten or Twenty? An Update on the Ongoing Debate on Gluten Safety Limits for Patients with Celiac Disease. GASTROINTESTINAL DISORDERS 2020; 2:202-211. [DOI: 10.3390/gidisord2030021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The only currently accepted and recommended treatment for individuals diagnosed with celiac disease (CD) is a strict life-long gluten-free diet (GFD). While the use of the GFD is well-established, strict adherence to diet is not easy to accomplish. In addition, the administration of a GFD may be compromised by inadvertent exposure to small amounts of gluten. International guidelines define a gluten-free product as one containing less than 20 parts per million (ppm), (20 milligrams of gluten per 1 kg of food) gluten. A number of reports have assessed the safe upper limit for gluten exposure for patients with CD, with general consensus that patients with CD should limit their daily intake to less than 50 mg.
Collapse
Affiliation(s)
- Inna Spector Cohen
- Pediatric Gastroenterology & Nutrition institute, Ruth Children’s Hospital of Haifa, Rambam Medical Center, Faculty of Medicine, Technion, Haifa 3109601, Israel
| | - Andrew Day
- Department of Paediatrics, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - Ron Shaoul
- Pediatric Gastroenterology & Nutrition institute, Ruth Children’s Hospital of Haifa, Rambam Medical Center, Faculty of Medicine, Technion, Haifa 3109601, Israel
| |
Collapse
|
42
|
Gaylord A, Trasande L, Kannan K, Thomas KM, Lee S, Liu M, Levine J. Persistent organic pollutant exposure and celiac disease: A pilot study. ENVIRONMENTAL RESEARCH 2020; 186:109439. [PMID: 32409013 DOI: 10.1016/j.envres.2020.109439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 06/11/2023]
Abstract
Celiac disease affects approximately 1% of the population worldwide. Little is known about environmental factors that may modulate risk in genetically susceptible populations. Persistent organic pollutants (POPs) are known endocrine disruptors and, given the interplay between the endocrine and immune systems, are plausible contributors to celiac disease. The current study aims to elucidate the association between POPs and celiac disease. We conducted a single-site pilot study of 88 patients recruited from NYU Langone's Hassenfeld Children's Hospital outpatient clinic, 30 of which were subsequently diagnosed with celiac disease using standard serology and duodenal biopsy examination. Polybrominated diphenyl ether (PBDEs), perfluoroalkyl substances (PFASs), and p,p'-dichlorodiphenyldichloroethylene (DDE) and HLA-DQ genotype category were measured in blood serum and whole blood, respectively. Multivariable logistic regressions were used to obtain odds ratios for celiac disease associated with serum POP concentrations. Controlling for sex, race, age, BMI, and genetic susceptibility score, patients with higher serum DDE concentrations had 2-fold higher odds of celiac disease (95% CI: 1.08, 3.84). After stratifying by sex, we found higher odds of celiac disease in females with serum concentrations of DDE (OR = 13.0, 95% CI = 1.54, 110), PFOS (OR = 12.8, 95% CI = 1.17, 141), perfluorooctanoic acid (OR = 20.6, 95% CI = 1.13, 375) and in males with serum BDE153, a PBDE congener (OR = 2.28, 95% CI = 1.01, 5.18). This is the first study to report on celiac disease with POP exposure in children. These findings raise further questions of how environmental chemicals may affect autoimmunity in genetically susceptible individuals.
Collapse
Affiliation(s)
- Abigail Gaylord
- Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Leonardo Trasande
- Department of Population Health, New York University School of Medicine, New York, NY, USA; Department of Pediatrics, New York University School of Medicine, USA; Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA; NYU Wagner School of Public Service, New York, NY, USA; NYU College of Global Public Health, New York, NY, USA
| | | | - Kristen M Thomas
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sunmi Lee
- NYU Medical Center, New York State Department of Health, Albany, NY, USA
| | - Mengling Liu
- Department of Population Health, New York University School of Medicine, New York, NY, USA; Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Jeremiah Levine
- Department of Pediatrics, New York University School of Medicine, USA; Division of Pediatric Gastroenterology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
43
|
Triolo TM, Pyle L, Seligova S, Yu L, Gottlieb PA, Steck AK. Risk of Islet and Celiac Autoimmunity in Cotwins of Probands With Type 1 Diabetes. J Endocr Soc 2020; 4:bvaa053. [PMID: 32537543 PMCID: PMC7278281 DOI: 10.1210/jendso/bvaa053] [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: 03/02/2020] [Accepted: 05/07/2020] [Indexed: 11/19/2022] Open
Abstract
Context Concordance for persistent islet autoimmunity (IA) and type 1 diabetes in monozygotic twins after probands are diagnosed is variable (30%-70%). Risk for development of IA in dizygotic twins is thought to be similar to nontwin siblings. Little is known in regard to the development of celiac autoimmunity (CDA) in twins of subjects with type 1 diabetes. Objective Our aim was to investigate the development of IA and CDA in cotwins of probands with type 1 diabetes. Methods Since 1995, the Twin Family Study has followed 336 twins (168 twin probands with type 1 diabetes and 168 cotwins) for a median of 14 years (interquartile range:10-18 years). Cotwins were followed for the development of IA, type 1 diabetes, and CDA. Results In monozygotic cotwins, cumulative incidence by age 20 was 14% for IA and 10% for CDA. Development of IA and CDA by age 20 was 9% and 12% in dizygotic cotwins, respectively. While the numbers are small, IA by age 30 years was 26% in monozygotic and 39% in dizygotic twins. In proportional hazards models, the proband’s younger age at diagnosis, but not sex or human leukocyte antigen were associated with time to IA and CDA in cotwins. Conclusion CDA risk by age 20 in cotwins was 10% to 12%. With long-term follow-up, cumulative incidence for IA is high in dizygotic twins, similar to monozygotic twins, suggesting a role of possible early environmental factors shared by type 1 diabetes discordant cotwins.
Collapse
Affiliation(s)
- Taylor M Triolo
- University of Colorado Anschutz Medical Campus - The Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Laura Pyle
- University of Colorado Anschutz Medical Campus, Pediatrics, Aurora, Colorado.,Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado
| | - Sona Seligova
- University of Colorado Anschutz Medical Campus - The Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Liping Yu
- University of Colorado Anschutz Medical Campus - The Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Peter A Gottlieb
- University of Colorado Anschutz Medical Campus - The Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Andrea K Steck
- University of Colorado Anschutz Medical Campus - The Barbara Davis Center for Diabetes, Aurora, Colorado
| |
Collapse
|
44
|
Pushing the Gluten-free Envelope: First Steps Towards Evidence-based Gluten-free Diet Recommendations. J Pediatr Gastroenterol Nutr 2020; 70:275-276. [PMID: 31899732 PMCID: PMC7144744 DOI: 10.1097/mpg.0000000000002614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
45
|
Choung RS, Khaleghi S, Cartee AK, Marietta EV, Larson JJ, King KS, Savolainen O, Ross AB, Rajkumar SV, Camilleri MJ, Rubio-Tapia A, Murray JA. Community-Based Study of Celiac Disease Autoimmunity Progression in Adults. Gastroenterology 2020; 158:151-159.e3. [PMID: 31560892 PMCID: PMC7065356 DOI: 10.1053/j.gastro.2019.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 09/06/2019] [Accepted: 09/16/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Celiac disease can develop at any age, but outcomes of adults with positive results from serologic tests for tissue transglutaminase antibodies (tTGA) without endoscopic determination of celiac disease (called celiac autoimmunity) have not been thoroughly evaluated. We investigated the proportion of adults with celiac autoimmunity at a community medical center and their progression to celiac disease. METHODS We analyzed waste blood samples from a community clinic from 15,551 adults for tTGA and, if titer results were above 2 U/mL, for endomysial antibody. The blood samples had been collected at 2 time points (median interval, 8.8 years) from 2006 through 2017. We collected data from the clinic on diagnoses of celiac disease based on duodenal biopsy analysis. RESULTS Of the serum samples collected at the first time point, 15,398 had negative results for tTGA, and 153 had positive results for tTGA (>4 U/mL). Based on medical records, 6 individuals received a diagnosis of celiac disease, for a cumulative incidence of celiac disease diagnosis of 0.06% (95% confidence interval, 0.01-0.11). Forty-nine (0.32%) individuals with a negative result from the first serologic test for tTGA had a positive result from the second test. Among the 153 adults who were tTGA positive at the first time point, 31 (20%) had a subsequent diagnosis of celiac disease, 81 (53%) remained positive for tTGA without a clinical diagnosis of celiac disease, and 41 (27%) had negative test results for tTGA at the second time point. Higher initial tTGA titers, female sex, and a history of hypothyroidism and autoimmune disease were associated with increased risks of subsequent diagnosis of celiac disease. Interestingly, adults whose first blood sample had a positive test result but second blood sample had a negative result for tTGA were older, had lower-than-average initial tTGA titer results, and had a higher mean body mass index than adults whose blood samples were positive for tTGA at both time points and adults later diagnosed with celiac disease. CONCLUSIONS In an analysis of serum samples collected from a community clinic an average of 8.8 years apart, we found that fewer than 1% of adults with negative results from an initial test for tTGA have a positive result on a second test. Of adults with positive results from the test for tTGA, only 20% are later diagnosed with celiac disease; the remaining individuals maintain persistent increases in tTGA without diagnoses of celiac disease or have negative results from second tests.
Collapse
Affiliation(s)
- Rok Seon Choung
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Shahryar Khaleghi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Amanda K Cartee
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Eric V Marietta
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Joseph J Larson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Katherine S King
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Otto Savolainen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Alastair B Ross
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Proteins and Metabolites Team, AgResearch, Lincoln, New Zealand
| | - S Vincent Rajkumar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael J Camilleri
- Department of Dermatology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Alberto Rubio-Tapia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
46
|
Autoimmune and Allergic Disorders are More Common in People With Celiac Disease or on a Gluten-free Diet in the United States. J Clin Gastroenterol 2019; 53:e416-e423. [PMID: 30045167 DOI: 10.1097/mcg.0000000000001100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
GOALS We analyzed demographics, lifestyle patterns, and clinical characteristics of people with celiac disease (CD) and people without CD avoiding gluten (PWAG) to better understand associations with medical conditions and consumer behavior. BACKGROUND Clinical significance of CD and gluten avoidance in the general population is incompletely understood. Recently, a high incidence of CD in adolescents with susceptibility genotypes, similar to other autoimmune or allergic disorders, and regional differences in consumer practices of gluten avoidance were reported. METHODS Among 22,277 participants in the National Health and Nutrition Examination Survey 2009-2014, we identified persons with CD by testing CD serology or by both a health care provider diagnosis and adherence to a gluten-free diet. Similarly, PWAG were defined as adherent to a gluten-free diet without a CD diagnosis. Consumer behavior and characteristics of both groups, CD and PWAG were compared with those without these conditions, using survey-weighted generalized logistic regression. RESULTS Participants with CD considered nutrition very important when grocery shopping and tended to have more constipation and thyroid disease. PWAG tended to spend more money on groceries, purchase organic foods, and check food labels more frequently during grocery shopping. They also reported having more food allergies, asthma, and thyroid disease. CONCLUSIONS Our study confirms that CD and PWAG share comorbidities of autoimmune nature. PWAG had more autoimmune/allergy-related disorders that may be associated with non-celiac gluten sensitivity a self-justifiable reason to be on the diet.
Collapse
|
47
|
Coburn S, Rose M, Streisand R, Sady M, Parker M, Suslovic W, Weisbrod V, Kerzner B, Kahn I. Psychological Needs and Services in a Pediatric Multidisciplinary Celiac Disease Clinic. J Clin Psychol Med Settings 2019; 27:433-443. [PMID: 31673859 DOI: 10.1007/s10880-019-09673-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Nellikkal SS, Hafed Y, Larson JJ, Murray JA, Absah I. High Prevalence of Celiac Disease Among Screened First-Degree Relatives. Mayo Clin Proc 2019; 94:1807-1813. [PMID: 31447136 DOI: 10.1016/j.mayocp.2019.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/24/2019] [Accepted: 03/04/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate the prevalence of first-degree relatives (FDRs) with celiac disease detected at screening and diagnostic significance of anti-tissue transglutaminase (anti-TTG). PATIENTS AND METHODS We performed a retrospective cohort study of 104 patients with a diagnosis of celiac disease and their FDRs, collecting data from electronic records of Mayo Clinic and celiac disease registry from December 20, 1983, to May 22, 2017. We collected demographics, presenting symptoms, indication for testing, family history, number of other family members screened, biopsy reports, and results of serologic tests. RESULTS Of 477 FDRs identified, 360 were screened (mean screening rate per family, 79%±25%) and 160 FDRs (44.4%) were diagnosed with celiac disease, at a mean age 31.9±21.6 years (62% female). All diagnosed FDRs had positive anti-TTG titers. Clinical features were documented in 148 diagnosed FDRs, of those 9 (6%) had classic, 97 (66%) had non-classic symptoms, and 42(28%) had no reported symptoms. Histology reports were available from 155 FDRs: 12 (8%) had Marsh 1, 77 (50%) had Marsh 3a, and 66 (43%) had Marsh 3b. A level of anti-TTG greater than or equal to 2.75 of the upper limit of normal identified FDRs with villous atrophy with 87% sensitivity, 82% specificity, and a positive predictive value of 95%. CONCLUSION In a retrospective cohort study of patients diagnosed with celiac disease, we found a high prevalence of celiac disease among screened FDRs. High anti-TTG titers associated with villous atrophy on small bowel biopsies, irrespective of symptoms.
Collapse
Affiliation(s)
- Shilpa S Nellikkal
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yamen Hafed
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Joseph J Larson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Imad Absah
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
49
|
Diagnosing Celiac Disease: Towards Wide-Scale Screening and Serology-Based Criteria? Gastroenterol Res Pract 2019; 2019:2916024. [PMID: 31467522 PMCID: PMC6701393 DOI: 10.1155/2019/2916024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022] Open
Abstract
Celiac disease is one of the most common food-related chronic disorders in children. Unfortunately, this multifaceted disease is challenging to recognize and remains markedly underdiagnosed. Screening of either known at-risk groups or even the whole population could increase the suboptimal diagnostic yield substantially. Many recent guidelines recommend screening of at least selected risk groups, but more wide-scale screening remains controversial. The increasing prevalence of celiac disease and the development of autoantibody assays have also led to a gradual shift in the diagnostics towards less invasive serology-based criteria in a subgroup of symptomatic children. The main open questions concern whether these criteria are applicable to all countries and clinical settings, as well as to adult patients. On the other hand, widening screening and the mistaken practice of initiating a gluten-free diet before the appropriate exclusion of celiac disease increase the number of borderline seropositive cases, which may also challenge the classical histopathological diagnostics. Sophisticated diagnostic methods and a deeper understanding of the natural history of early developing celiac disease may prove useful in these circumstances.
Collapse
|
50
|
Abstract
OBJECTIVES To determine the association between the amount of gluten intake in childhood and later celiac disease (CD), for which data are currently scarce. METHODS The prospective Diabetes Autoimmunity Study in the Young cohort includes 1875 at-risk children with annual estimates of gluten intake (grams/d) from age 1 year. From 1993 through January 2017, 161 children, using repeated tissue transglutaminase (tTGA) screening, were identified with CD autoimmunity (CDA) and persistent tTGA positivity; of these children, 85 fulfilled CD criteria of biopsy-verified histopathology or persistently high tTGA levels. Cox regression, modeling gluten intake between ages 1 and 2 years (i.e., in 1-year-olds), and joint modeling of cumulative gluten intake throughout childhood were used to estimate hazard ratios adjusted for confounders (aHR). RESULTS Children in the highest third of gluten intake between the ages of 1 and 2 years had a 2-fold greater hazard of CDA (aHR 2.17; 95% confidence interval [CI], 1.22-3.88; P value = 0.01) and CD (aHR 1.96; 95% CI, 0.90-4.24; P value = 0.09) than those in the lowest third. The risk of developing CDA increased by 5% per daily gram increase in gluten intake (aHR 1.05; 95% CI, 1.00-1.09; P value = 0.04) in 1-year-olds. The association between gluten intake in 1-year-olds and later CDA or CD did not differ by the child's human leukocyte antigen genotype. The incidence of CD increased with increased cumulative gluten intake throughout childhood (e.g., aHR 1.15 per SD increase in cumulative gluten intake at age 6; 95% CI, 1.00-1.32; P value = 0.04). DISCUSSION Gluten intake in 1-year-olds is associated with the future onset of CDA and CD in children at risk for the disease.
Collapse
|