1
|
Szajewska H, Shamir R, Auricchio R, Chmielewska A, Dolinsek J, Kivelä L, Koletzko S, Korponay-Szabo IR, Af Segerstad EMH, Mearin ML, Meijer-Boekel C, Konickx CR, Rodriguez-Herrera A, Stordal K, Troncone R, Wessels M. Early diet and the risk of coeliac disease. An update 2024 position paper by the ESPGHAN special interest group on coeliac disease. J Pediatr Gastroenterol Nutr 2024; 79:438-445. [PMID: 38847232 DOI: 10.1002/jpn3.12280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 08/03/2024]
Abstract
This position paper by the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) Special Interest Group on Coeliac Disease (SIG-CD) presents an update to the 2016 recommendations concerning early diet and the risk of coeliac disease (CD). This update adheres to the policy that mandates reviewing guidelines every 5 years, particularly when new data emerge. The 2024 statements and recommendations are essentially similar to the 2016 recommendations. Breastfeeding, whether any amount, exclusive, or of any duration, does not reduce the risk of developing CD. Introducing gluten into an infant's diet at any time between completed 4 months (≥17 weeks) and 12 months of age does not affect the cumulative incidence of CD, although earlier introduction may lead to earlier seroconversion and CD. In observational studies involving cohorts with a known risk for CD, consuming a high amount of gluten compared to a low amount during weaning and in the subsequent childhood years-specifically the first 2-3 years, and even up to 5 years in some studies-was associated with an increased risk for CD. However, the specific optimal amounts of gluten consumption remain undetermined due to insufficient evidence on safe thresholds, and the impact of restricting gluten in the diet of healthy children of unknown risk for CD is unknown. Thus, any recommendation on the gluten amount is currently unjustifiable for the general population and infants with known HLA risk types. There is no specific guidance on the type of gluten-containing foods to be introduced at weaning.
Collapse
Affiliation(s)
- Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Raanan Shamir
- Institute of Gastroenterology, Nutrition and Liver Disease, Schneider Children's Medical Center of Israel, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Renata Auricchio
- Department of Translational Medical Sciences and European Laboratory for the Investigation of Food Induced Diseases, University Federico II, Naples, Italy
| | - Anna Chmielewska
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Jernej Dolinsek
- Department of Pediatrics, University Medical Center Maribor, Maribor, Slovenia
| | - Laura Kivelä
- Celiac Disease Research Center, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland
- Department of Paediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Munich, Germany
- Department of Pediatrics, School of Medicine Collegium Medicum, Gastroenterology and Nutrition, University of Warmia and Mazury, Olsztyn, Poland
| | - Ilma R Korponay-Szabo
- Celiac Disease Center, Heim Pál National Paediatric Institute, Budapest, Hungary
- Department of Paediatrics, University of Debrecen, Debrecen, Hungary
| | - Elin M Hård Af Segerstad
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Unit of Celiac and Diabetes Research, Clinical Sciences Malmö, Lund university, Lund, Sweden
- Department of Pediatrics, Skane University Hospital, Malmö, Sweden
| | - M Luisa Mearin
- Department of Paediatric Gastroenterology, Leiden University Medical Center - Willem Alexander Children's Hospital, Leiden, The Netherlands
| | - Caroline Meijer-Boekel
- Department of Paediatric Gastroenterology, Leiden University Medical Center - Willem Alexander Children's Hospital, Leiden, The Netherlands
| | - Carmen Ribes Konickx
- Pediatric Gastroenterology, La Fe University Hospital, Instituto de Investigation Sanitaria La FE Valencia, Valencia, Spain
| | | | - Ketil Stordal
- Department of Paediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Riccardo Troncone
- Department of Translational Medical Sciences and European Laboratory for the Investigation of Food Induced Diseases, University Federico II, Naples, Italy
| | - Margreet Wessels
- Department of Paediatrics, Rijnstate Hospital, Arnhem, The Netherlands
| |
Collapse
|
2
|
Lungaro L, Costanzini A, Manza F, Caputo F, Remelli F, Volpato S, De Giorgio R, Volta U, Caio G. Celiac Disease: A Forty-Year Analysis in an Italian Referral Center. Nutrients 2024; 16:2292. [PMID: 39064734 PMCID: PMC11279732 DOI: 10.3390/nu16142292] [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: 06/25/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Celiac disease (CD) is an autoimmune disorder triggered by gluten ingestion. Herein, we assessed clinical, serological and histopathological findings of a single-center, large cohort of CD patients diagnosed and followed-up over forty years. METHODS From January 1980 to December 2020, 1547 CD patients (1170 females; age range: 8-81 years; F:M ratio = 3.1:1) were diagnosed in an Italian tertiary referral center. Comorbidities and complications were recorded at diagnosis and during follow-up. RESULTS CD diagnoses quadrupled after 2000. The most frequent phenotype was the non-classical CD (63.3%), and the most prevalent histotype was Marsh 3C (44.7%). Gastrointestinal manifestations, detectable in 51% of patients, were diarrhea (24.3%), bloating (28%) and aphthous stomatitis (19.7%). The most common CD-associated disorder was osteopenia (59.9%), predominant in females (64.3%); extraintestinal manifestations included anemia (35.8% iron-deficiency; 87% folic acid malabsorption), cryptogenic hypertransaminasemia (27.9%), and recurrent miscarriages (11.5%). Thyroiditis (26.9%), type 1 diabetes mellitus (2.9%), and dermatitis herpetiformis (1.4%) were the most common CD-related autoimmune disorders. Six patients had inflammatory bowel disease. Complications and mortality rate occurred in 1.8% and 1.9%, respectively. CONCLUSIONS This single-center, large cohort analysis confirmed that CD presentation changed over the years, with an increase of non-classical and subclinical clinical phenotypes.
Collapse
Affiliation(s)
- Lisa Lungaro
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
| | - Anna Costanzini
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
| | - Francesca Manza
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
| | - Fabio Caputo
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
| | - Francesca Remelli
- Department of Medical Science, University of Ferrara, 44124 Ferrara, Italy; (F.R.); (S.V.)
| | - Stefano Volpato
- Department of Medical Science, University of Ferrara, 44124 Ferrara, Italy; (F.R.); (S.V.)
- Geriatrics Unit, Azienda Ospedaliero-Universitaria of Ferrara, 44124 Ferrara, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
| | - Umberto Volta
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Giacomo Caio
- Department of Translational Medicine, St. Anna Hospital, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (A.C.); (F.M.); (F.C.)
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital-Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
3
|
Galipeau HJ, Hinterleitner R, Leonard MM, Caminero A. Non-Host Factors Influencing Onset and Severity of Celiac Disease. Gastroenterology 2024; 167:34-50. [PMID: 38286392 DOI: 10.1053/j.gastro.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts; Center for Celiac Research and Treatment, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
4
|
Lindgren M, Palmkvist E, Norström F, Cerqueiro Bybrant M, Myleus A, Samuelsson U, Ludvigsson J, Carlsson A. Cumulative incidence of type 1 diabetes in two cohorts of children with different national gluten recommendations in infancy. Acta Diabetol 2024; 61:35-41. [PMID: 37589890 PMCID: PMC10806042 DOI: 10.1007/s00592-023-02168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/15/2023] [Indexed: 08/18/2023]
Abstract
AIMS Between 1985 and 1996, Sweden experienced an "epidemic" of celiac disease with a fourfold increase in incidence in young children. Timing and amount of gluten introduced during infancy have been thought to explain this "epidemic". We aimed to study whether the cumulative incidence of type 1 diabetes differs between children born during the "epidemic" compared to children born after. METHODS This is a national register study in Sweden comparing the cumulative incidence of type 1 diabetes in two birth cohorts of 240 844 children 0-17 years old born 1992-1993, during the "epidemic", and 179 530 children born 1997-1998, after the "epidemic". Children diagnosed with type 1 diabetes were identified using three national registers. RESULTS The cumulative incidence of type 1 diabetes by the age of 17 was statistically significantly higher in those born after the "epidemic" 0.77% than in those born during the "epidemic" 0.68% (p < 0.001). CONCLUSION The incidence of type 1 diabetes is higher in those born after the epidemic compared to those born during the epidemic, which does not support the hypothesis that gluten introduction increases the incidence of T1D. Changes in gluten introduction did not halt the increased incidence of type 1 diabetes in Sweden.
Collapse
Affiliation(s)
- Marie Lindgren
- Department of Clinical Science, Lund University, Lund, Sweden.
- Children's Clinic, Vrinnevi Hospital, Norrköping, Sweden.
| | - Elsa Palmkvist
- Department of Clinical Science, Lund University, Lund, Sweden
| | - Fredrik Norström
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
| | - Mara Cerqueiro Bybrant
- Paediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Anna Myleus
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Ulf Samuelsson
- Crown Princess Victoria's Children's Hospital, Region Östergötland, Linköping, Sweden
- Division of Paediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria's Children's Hospital, Region Östergötland, Linköping, Sweden
- Division of Paediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Annelie Carlsson
- Department of Clinical Science, Lund University, Lund, Sweden
- Department of Pediatric, Skånes University hospital, Lund, Sweden
| |
Collapse
|
5
|
Olshan KL, Leonard MM. Nutrition and risk of celiac disease - you are what you (wh)eat. Am J Clin Nutr 2023; 118:1071-1072. [PMID: 38044021 DOI: 10.1016/j.ajcnut.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/18/2023] [Indexed: 12/05/2023] Open
Affiliation(s)
- Katherine L Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, United States; Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, United States; Harvard Medical School, Celiac Education and Research Program, Boston, MA, United States
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, United States; Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, United States; Harvard Medical School, Celiac Education and Research Program, Boston, MA, United States.
| |
Collapse
|
6
|
Crawley C, Sander SD, Nohr EA, Nybo Andersen AM, Husby S. Early environmental risk factors and coeliac disease in adolescents: a population-based cohort study in Denmark. BMJ Open 2023; 13:e061006. [PMID: 38011980 PMCID: PMC10685961 DOI: 10.1136/bmjopen-2022-061006] [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: 01/13/2022] [Accepted: 10/19/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVES Our aim was to investigate the association between early environmental factors and the development of coeliac disease (CeD) in adolescents, recruited from a cohort nested in the Danish National Birth Cohort (DNBC). DESIGN The study was designed as a prospective cohort study, nested in DNBC PARTICIPANTS: The Glutenfunen cohort comprises 1266 participants, nested in DNBC. All participants were screened for CeD, and in total, 28 cases of biopsy proven CeD were identified. Data about breastfeeding, timing of introduction to solid food in infancy, use of antibiotics, infections and symptoms were parentally reported prospectively at 6 months and 18 months, respectively. We estimated ORs and 95% CIs of CeD in adolescents using logistic regression analysis. RESULTS Viral croup reported at 18 months of age was associated with CeD in adolescents with an OR of 3.2 (95% CI: 1.2 to 8.7). Furthermore, otitis media also reported at 18 months of age was linked with CeD with an OR of 3.2 (95% CI: 1.5 to 7.3). We were not able to find any statistical associations between CeD and breastfeeding, frequency of infections, parentally reported use of antibiotic and timing of solid foods. CONCLUSION In this study, we present an overview of the relationship between early environmental factors and occurrence of CeD in adolescents. Our findings, despite limitations due to a limited number of cases of CeD, suggest a role of viral infections in the pathogenesis of CeD.
Collapse
Affiliation(s)
- Cæcilie Crawley
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Hans Christian Anderson Children's Hospital, Odense University Hospital, Odense, Denmark
| | | | - Ellen Aagaard Nohr
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Research Unit of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| | | | - Steffen Husby
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Hans Christian Anderson Children's Hospital, Odense University Hospital, Odense, Denmark
| |
Collapse
|
7
|
Kazakova P, Abasolo N, de Cripan SM, Marquès E, Cereto-Massagué A, Garcia L, Canela N, Tormo R, Torrell H. Gut Microbiome and Small RNA Integrative-Omic Perspective of Meconium and Milk-FED Infant Stool Samples. Int J Mol Sci 2023; 24:ijms24098069. [PMID: 37175775 PMCID: PMC10179101 DOI: 10.3390/ijms24098069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The human gut microbiome plays an important role in health, and its initial development is conditioned by many factors, such as feeding. It has also been claimed that this colonization is guided by bacterial populations, the dynamic virome, and transkingdom interactions between host and microbial cells, partially mediated by epigenetic signaling. In this article, we characterized the bacteriome, virome, and smallRNome and their interaction in the meconium and stool samples from infants. Bacterial and viral DNA and RNA were extracted from the meconium and stool samples of 2- to 4-month-old milk-fed infants. The bacteriome, DNA and RNA virome, and smallRNome were assessed using 16S rRNA V4 sequencing, viral enrichment sequencing, and small RNA sequencing protocols, respectively. Data pathway analysis and integration were performed using the R package mixOmics. Our findings showed that the bacteriome differed among the three groups, while the virome and smallRNome presented significant differences, mainly between the meconium and stool of milk-fed infants. The gut environment is rapidly acquired after birth, and it is highly adaptable due to the interaction of environmental factors. Additionally, transkingdom interactions between viruses and bacteria can influence host and smallRNome profiles. However, virome characterization has several protocol limitations that must be considered.
Collapse
Affiliation(s)
- Polina Kazakova
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | - Nerea Abasolo
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | - Sara Martinez de Cripan
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | | | - Adrià Cereto-Massagué
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | - Lorena Garcia
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | - Núria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| | - Ramón Tormo
- ESPGHAN, European Society for Paediatric Gastroenterology, Hepatology and Nutrition, 1201 Geneva, Switzerland
- Gastroenterology and Nutrition Pediatric Center, 08006 Barcelona, Spain
| | - Helena Torrell
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), 43204 Reus, Spain
| |
Collapse
|
8
|
Thurkkada AP, Rajasekharan Nair S, Thomas S, Sreelekha P, Sanu SK, Chandran PR, Pillai Sreekanth G. Effectiveness of Hoffman's Exercise in Postnatal Mothers With Grade 1 Inverted Nipples. J Hum Lact 2023; 39:69-75. [PMID: 35695389 DOI: 10.1177/08903344221102890] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Breastfeeding provides perfect nutrition for infants. The inverted nipples in mothers make breastfeeding more challenging. Besides surgical approaches, non-pharmacological interventions are also gaining importance. RESEARCH AIM To evaluate the effectiveness of Hoffman's exercise on breastfeeding among postnatal mothers with grade I inverted nipples. METHOD A quantitative approach with a parallel arm randomized controlled trial was used in the study. Postnatal mothers were screened for inverted nipples using a pinch test. Participants with at least one inverted nipple (Grade 1) were randomly allocated into two groups: the experimental group (supported with Hoffman's exercise; n = 28, 50.9% ) and the control group (without Hoffman's exercise; n = 27, 49.1%). The pre-test data, including demographics, nipple length, and breastfeeding assessment, were collected. Breastfeeding assessment was evaluated using the Bristol Breastfeeding Assessment Tool (BBAT). On the 3rd day, the post-test data, including nipple length and Breastfeeding Assessment, were collected among the groups. RESULTS The BBAT assessment was significantly higher in the post-test compared to that of the pre-test in the participants provided with Hoffman's exercise. The nipple length was found to be higher in participants provided with Hoffman's exercise. Furthermore, the variables-including age, gravida, nipple problems, and delivery type-were not found to have any significant effect with either pre-test or post-test levels of breastfeeding. CONCLUSIONS Hoffman's exercise was found to be an effective method to improve breastfeeding in Grade 1 nipple-inverted among post-natal mothers. This nipple exercise is inexpensive, easy to follow, and results in the successful initiation of breastfeeding. CLINICAL TRIAL REGISTRY AND REGISTRATION NUMBER CTRI/2019/05/019279, May 23, 2019 (retrospectively registered).
Collapse
Affiliation(s)
| | | | - Sara Thomas
- Amrita College of Nursing, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Parvathy Sreelekha
- Amrita College of Nursing, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | - Parvathy R Chandran
- CSIR- National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
| | - Gopinathan Pillai Sreekanth
- Siriraj Center of Research Excellence for Molecular Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| |
Collapse
|
9
|
Alotiby AA. The role of breastfeeding as a protective factor against the development of the immune-mediated diseases: A systematic review. Front Pediatr 2023; 11:1086999. [PMID: 36873649 PMCID: PMC9981158 DOI: 10.3389/fped.2023.1086999] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
INTRODUCTION Breast milk is rich in nutrients and immunological factors capable of protecting infants against various immunological diseases and disorders. The current systematic review has been framed with the objective of studying the role of breastfeeding as a protective factor against the development of immune-mediated diseases. METHODS The database and website searches were performed using PubMed, PubMed Central, Nature, Springer, Nature, Web of Science, and Elsevier. The studies were scrutinized based on the nature of participants and the nature of disease considered. The search was restricted to infants with immune-mediated diseases such as diabetes mellitus, allergic conditions, diarrhoea, and rheumatoid arthritis. RESULTS We have included 28 studies, out of which seven deal with diabetes mellitus, two rheumatoid arthritis, five studies about Celiac Disease, twelve studies about allergic/ asthma/wheezing conditions and one study on each of the following diseases: neonatal lupus erythematosus and colitis. DISCUSSION Based on our analysis, breastfeeding in association with the considered diseases was found to be positive. Breastfeeding is involved as protective factor against various diseases. The role of breastfeeding in the prevention of diabetes mellitus has been found to be significantly higher than for other diseases.
Collapse
Affiliation(s)
- Amna A Alotiby
- Department of Hematology and Immunology, Faculty of Medicine Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
10
|
Lal SB, Venkatesh V, Aneja A, Seetharaman K, Kumar Y, Prasad KK, Rana SS. Clinical spectrum & changing presentation of celiac disease in Indian children. Indian J Med Res 2023; 158:75-84. [PMID: 37602589 PMCID: PMC10550060 DOI: 10.4103/ijmr.ijmr_1102_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 08/10/2023] Open
Abstract
Background & objectives Celiac disease (CD) is a genetic immune mediated disorder characterised by gluten intolerance. This single centre study, from north India was aimed to assess the clinical, serological and histological profile of CD in a large cohort of children and the changing trends in its presentation. Methods A review of clinical details of CD children diagnosed between 2000 and 2019 and currently on follow up was performed. Information on demography, symptoms, associated conditions, serology, biopsy findings and gluten-free diet were analyzed. Results The mean age (±standard deviation) of 891 children included in the study, at onset and at diagnosis was 4.0±2.7 and 6.2±3.1 yr, respectively. Growth faltering, abdominal pain, abdominal distension and diarrhoea were presenting symptoms in 70, 64.2, 61.2 and 58.2 per cent, respectively. A positive family history of CD was present in 14 per cent and autoimmune conditions in 12.3 per cent of children. Thyroid disorders were seen in 8.5 per cent of children and Type 1 diabetes mellitus (T1DM) in 5.7 per cent. The duration of breastfeeding had a weak positive correlation with age at onset and diagnosis of CD (P<0.001). Non-classical CD was significantly more common in children aged >10 yr and in those presenting after 2010 (P<0.01). T1DM and hypothyroidism occurred more frequently in non-compliant children. Interpretation & conclusions This was the largest single centre study, pertaining to the presentation and follow up of CD in children. Infants and young children were more likely to present with classical symptoms of diarrhoea, abdominal distension and growth failure while older children presented with non-classical CD. There was a trend towards non-classical forms of CD in recent years.
Collapse
Affiliation(s)
- Sadhna Bhasin Lal
- Division of Paediatric Gastroenterology & Hepatology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Vybhav Venkatesh
- Division of Paediatric Gastroenterology & Hepatology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Aradhana Aneja
- Division of Paediatric Gastroenterology & Hepatology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Keerthivasan Seetharaman
- Division of Paediatric Gastroenterology & Hepatology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Yashwant Kumar
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Kaushal Kishor Prasad
- Division of Gastroenterology Histopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Surinder Singh Rana
- Department of Gastroenterology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
11
|
Szajewska H, Shamir R, Stróżyk A, Chmielewska A, Zalewski BM, Auricchio R, Koletzko S, Korponay-Szabo IR, Mearin ML, Meijer C, Ribes-Koninckx C, Troncone R. Systematic review: early feeding practices and the risk of coeliac disease. A 2022 update and revision. Aliment Pharmacol Ther 2023; 57:8-22. [PMID: 36411726 DOI: 10.1111/apt.17290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The effects of early feeding practices on the risk of coeliac disease (CD) remain debated. AIMS To update evidence on these practices on the risk of CD and/or CD-related autoimmunity (CDA), defined as anti-transglutaminase or anti-endomysial antibody positivity METHODS: We searched MEDLINE, EMBASE and the Cochrane Library to May 2022 for randomised controlled trials (RCTs) and observational studies. RESULTS We included 36 publications (30 studies). In the population at genetic risk of developing CD (HLA DQ2/DQ8-positive), exclusive or any breastfeeding and longer breastfeeding duration did not reduce the risk of developing CD/CDA during childhood. While a meta-analysis of four case-control studies showed a decreased risk for CD when gluten was introduced during breastfeeding, this was not shown in RCTs and cohort studies. Age at gluten introduction was not associated with cumulative CD/CDA risk, although two RCTs suggested that earlier gluten introduction was associated with earlier CDA appearance. Evidence from six observational studies suggests that consumption of a higher amount of gluten at weaning and/or thereafter may increase CD risk. There is insufficient evidence to determine the amount of gluten associated with an increased CD/CDA risk. Regarding whether infant feeding practices modulate the risk conferred by different HLA genotypes results were inconsistent. CONCLUSIONS For the population at genetic risk of CD, breastfeeding and age at gluten introduction have no effect on its cumulative incidence during childhood. There is some evidence for an effect of the amount of gluten consumed at weaning and/or thereafter on CD/CDA risk.
Collapse
Affiliation(s)
- Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Raanan Shamir
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Agata Stróżyk
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Anna Chmielewska
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | | | - Renata Auricchio
- Department of Translation Medical Science, Pediatric Section European Laboratory for the Investigation of Food Induced Disease (ELFID), University Federico II, Naples, Italy
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany
- School of Medicine Collegium Medicum, Department of Pediatrics, Gastroenterology and Nutrition, University of Warmia and Mazury, Olsztyn, Poland
| | - Ilma R Korponay-Szabo
- Department of Pediatrics, Faculty of Medicine and Clinical Center, University of Debrecen, Debrecen, Hungary
- Celiac Disease Center, Heim Pál National Paediatric Institute, Budapest, Hungary
| | - M Luisa Mearin
- Willem Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Caroline Meijer
- Willem Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Carmen Ribes-Koninckx
- Pediatric Gastroenterology and Hepatology & Instituto de Investigacion Sanitaria, La Fe University Hospital, Valencia, Spain
| | - Riccardo Troncone
- Department of Translation Medical Science, Pediatric Section European Laboratory for the Investigation of Food Induced Disease (ELFID), University Federico II, Naples, Italy
| |
Collapse
|
12
|
Hård af Segerstad EM, Liu X, Uusitalo U, Agardh D, Andrén Aronsson C. 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] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [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
|
13
|
Chang D, O’Shea D, Therrien A, Silvester JA. Review article: Becoming and being coeliac-special considerations for childhood, adolescence and beyond. Aliment Pharmacol Ther 2022; 56 Suppl 1:S73-S85. [PMID: 35815825 PMCID: PMC9441244 DOI: 10.1111/apt.16851] [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: 01/08/2022] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 12/09/2022]
Abstract
Classically considered a disease of early childhood characterised by malabsorption and failure to thrive, coeliac disease is now recognised to arise in genetically susceptible individuals at any age. Although permissive HLA genotypes are the strongest predictor of coeliac disease, they are not sufficient. Several prospective cohort studies enrolling genetically at-risk infants have investigated the role of potential triggers of coeliac disease autoimmunity, such as timing of gluten introduction, viral infections and dietary patterns. Much less is known about triggers of coeliac disease in adulthood. Better understanding of factors leading to coeliac disease may be helpful in the management of those with potential coeliac disease (elevated serum celiac antibodies without villous atrophy in the small intestine), many of whom initiate a gluten-free diet without demonstration of villous atrophy. There are a range of clinical presentations of celiac disease in childhood and patterns of coeliac serology, including fluctuation and spontaneous reversion on a gluten-containing diet, vary. There is a current debate over best strategies to manage adults and children with potential coeliac disease to avoid over-treatment and under-treatment. Childhood and adolescence carry unique issues pertaining to the diagnosis and management of coeliac disease, and include nutrition and growth, rescreening, repeat biopsy, dietary adherence concerns and transition to adult care. In conclusion, while coeliac disease has similar pathogenesis and general clinical manifestations in paediatric and adult populations, diagnostic and management approaches need to adapt to the developmental stages.
Collapse
Affiliation(s)
- Denis Chang
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA
| | - Delia O’Shea
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA
| | - Amelie Therrien
- 2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA,3. Beth Israel Deaconess Medical Center, Boston, MA
| | - Jocelyn A Silvester
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA,3. Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
14
|
Makharia GK, Chauhan A, Singh P, Ahuja V. Review article: Epidemiology of coeliac disease. Aliment Pharmacol Ther 2022; 56 Suppl 1:S3-S17. [PMID: 35815830 DOI: 10.1111/apt.16787] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022]
Abstract
Coeliac disease is an immune-mediated disease caused by ingestion of gluten in genetically susceptible individuals. Coeliac disease has been thought to affect mainly people of European origin but subsequently many studies revealed that it affects people living in North America, Oceania, South America, Asia as well as Africa. The global pooled seroprevalence and prevalence of biopsy-confirmed coeliac disease are 1.4% and 0.7% respectively. The pooled incidence rates in women and men are 17.4 (95% CI: 13.7-21.1) and 7.8 (95% CI: 6.3-9.2) per 100 000 person-years respectively. The systematic reviews, based on many population-based data, suggest that both the prevalence and the incidence of coeliac disease has increased over past three decades, which may be attributable not only to an increase in the detection rate (improvement in diagnostic tests, simplification of diagnostic criteria and increase in awareness about the disease) but also because of modernisation and globalisation related changes in the dietary practices including increase in the use of convenience food and dietary gluten. In addition to genetic factors, while there are many environmental risk factors, including age at the first introduction of gluten, breastfeeding, caesarean section, exposure to antibiotics and gut microbiome; the amount of gluten ingestion during early part of life, however, has been shown to increase the risk of coeliac disease, and this is relevant from the point of view of primary prevention. In this review, we have reviewed and summarised the literature, up till year 2021, related to the global and continent-wise epidemiology and risk factors associated with coeliac disease.
Collapse
Affiliation(s)
- Govind K Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Chauhan
- Department of Gastroenterology, Indira Gandhi Medical College and Hospital, Shimla, India
| | - Prashant Singh
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Vineet Ahuja
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
15
|
Felber J, Bläker H, Fischbach W, Koletzko S, Laaß M, Lachmann N, Lorenz P, Lynen P, Reese I, Scherf K, Schuppan D, Schumann M, Aust D, Baas S, Beisel S, de Laffolie J, Duba E, Holtmeier W, Lange L, Loddenkemper C, Moog G, Rath T, Roeb E, Rubin D, Stein J, Török H, Zopf Y. Aktualisierte S2k-Leitlinie Zöliakie der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS). ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:790-856. [PMID: 35545109 DOI: 10.1055/a-1741-5946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Jörg Felber
- Medizinische Klinik II - Gastroenterologie, Hepatologie, Endokrinologie, Hämatologie und Onkologie, RoMed Klinikum Rosenheim, Rosenheim, Deutschland
| | - Hendrik Bläker
- Institut für Pathologie, Universitätsklinikum Leipzig AöR, Leipzig, Deutschland
| | | | - Sibylle Koletzko
- Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU-Klinikum München, München, Deutschland.,Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, 10-719 Olsztyn, Polen
| | - Martin Laaß
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland
| | - Nils Lachmann
- Institut für Transfusionsmedizin, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Pia Lorenz
- Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
| | - Petra Lynen
- Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
| | - Imke Reese
- Ernährungsberatung und -therapie Allergologie, München, Deutschland
| | - Katharina Scherf
- Institute of Applied Biosciences Department of Bioactive and Functional Food Chemistry, Karlsruhe Institute of Technology (KIT), Karlsruhe, Deutschland
| | - Detlef Schuppan
- Institut für Translationale Immunologie, Johannes Gutenberg-Universität Mainz, Mainz, Deutschland.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael Schumann
- Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Deutschland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
The global burden of coeliac disease: opportunities and challenges. Nat Rev Gastroenterol Hepatol 2022; 19:313-327. [PMID: 34980921 DOI: 10.1038/s41575-021-00552-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Coeliac disease is a systemic disorder characterized by immune-mediated enteropathy, which is caused by gluten ingestion in genetically susceptible individuals. The clinical presentation of coeliac disease is highly variable and ranges from malabsorption through solely extra-intestinal manifestations to asymptomatic. As a result, the majority of patients with coeliac disease remain undiagnosed, misdiagnosed or experience a substantial delay in diagnosis. Coeliac disease is diagnosed by a combination of serological findings of disease-related antibodies and histological evidence of villous abnormalities in duodenal biopsy samples. However, variability in histological grading and in the diagnostic performance of some commercially available serological tests remains unacceptably high and confirmatory assays are not readily available in many parts of the world. Currently, the only effective treatment for coeliac disease is a lifelong, strict, gluten-free diet. However, many barriers impede patients' adherence to this diet, including lack of widespread availability, high cost, cross-contamination and its overall restrictive nature. Routine follow-up is necessary to ensure adherence to a gluten-free diet but considerable variation is evident in follow-up protocols and the optimal disease management strategy is not clear. However, these challenges in the diagnosis and management of coeliac disease suggest opportunities for future research.
Collapse
|
17
|
Flores Monar GV, Islam H, Puttagunta SM, Islam R, Kundu S, Jha SB, Rivera AP, Sange I. Association Between Type 1 Diabetes Mellitus and Celiac Disease: Autoimmune Disorders With a Shared Genetic Background. Cureus 2022; 14:e22912. [PMID: 35399440 PMCID: PMC8986520 DOI: 10.7759/cureus.22912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 11/05/2022] Open
|
18
|
Treppiccione L, Luongo D, Maurano F, Rossi M. Next generation strategies to recover immunological tolerance in celiac disease. Int Rev Immunol 2022; 42:237-245. [PMID: 35225129 DOI: 10.1080/08830185.2022.2044807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Celiac disease (CD) is an autoimmune disease that occurs in genetically predisposed individuals following the ingestion of gluten. Its prevalence is rising worldwide. A gluten-free (GF) diet is mandatory for the management of CD. However, several issues persist regarding the nutritional quality of GF products. Importantly, deep knowledge about the pathogenic mechanisms in CD highlights the central role of CD4+ T cell-mediated immunity in CD. Furthermore, intestinal T regulatory cells are functional in CD, but cytokines such as IL-15, produced under inflammatory conditions, hamper their activity. This paves the way for the development of immunomodulatory strategies to the GF diet. From this perspective, microbiological approaches were considered able to modulate the gluten-specific immune response. Interestingly, gliadin peptide-based immunotherapy to abolish the inflammatory CD4+T cell-mediated response has been explored in CD patients. Furthermore, different biotechnological approaches based on the use of chemically/enzymatically modified gluten molecules have been proved effective in different models of CD. However, the choice of the right age in infants to introduce the antigen and thus induce tolerance still remains an important issue to solve. Addressing all these points should help to design an effective intervention strategy for preventing CD.
Collapse
Affiliation(s)
| | | | | | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy
| |
Collapse
|
19
|
Ali B, Khan AR. Efficacy of Probiotics in Management of Celiac Disease. Cureus 2022; 14:e22031. [PMID: 35340497 PMCID: PMC8912170 DOI: 10.7759/cureus.22031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 11/05/2022] Open
|
20
|
Associations Between Subclass Profile of IgG Response to Gluten and the Gastrointestinal and Motor Symptoms in Children With Cerebral Palsy. J Pediatr Gastroenterol Nutr 2021; 73:367-375. [PMID: 34231978 PMCID: PMC8380641 DOI: 10.1097/mpg.0000000000003181] [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] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Gastrointestinal problems are often seen in children with cerebral palsy, although the etiology and underlying mechanisms are not fully understood. Recent data point to significantly elevated levels of IgG antibody to dietary gluten in cerebral palsy independent of celiac disease, a gluten-mediated autoimmune enteropathy. We aimed to further characterize this antibody response by examining its subclass distribution and target reactivity in the context of relevant patient symptom profile. METHODS Study participants included children with cerebral palsy (n = 70) and celiac disease (n = 85), as well as unaffected controls (n = 30). Serum IgG antibody to gluten was investigated for subclass distribution, pattern of reactivity towards target proteins, and relationship with gastrointestinal symptoms and motor function. RESULTS The anti-gluten IgG antibody response in the cerebral palsy cohort was constituted of all 4 subclasses. In comparison with celiac disease, however, IgG1, IgG2, and IgG3 subclasses were significantly lower, whereas the IgG4 response was significantly higher in cerebral palsy. Within the cohort of cerebral palsy patients, levels of anti-gluten IgG1, IgG3, and IgG4 were greater in those with gastrointestinal symptoms, and the IgG3 subclass antibody correlated inversely with gross motor function. The anti-gluten IgG antibodies targeted a broad range of gliadin and glutenin proteins. CONCLUSIONS These findings reveal an anti-gluten IgG subclass distribution in cerebral palsy that is significantly different from that in celiac disease. Furthermore, the observed association between IgG subclass and symptom profile is suggestive of a relationship between the immune response and disease pathophysiology that may indicate a role for defects in gut immune and barrier function in cerebral palsy.
Collapse
|
21
|
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: 4.3] [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
|
22
|
Llorente Pelayo S, Palacios Sánchez M, Docio Pérez P, Gutiérrez Buendía D, Peña Sainz-Pardo E, Vega Santa-Cruz B, García Calatayud S. Infections in early life as risk factor for coeliac disease. ANALES DE PEDIATRÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.anpede.2020.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
23
|
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
|
24
|
Lomash A, Venkatakrishnan A, Bothra M, Dhingra B, Kumar P, Kapoor S. Coeliac disease presenting atypically: A much wider spectrum. Trop Doct 2021; 51:309-314. [PMID: 33573512 DOI: 10.1177/0049475521991348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atypical coeliac disease in young children is frequently missed when it presents atypically as non-gastrointestinal presentations to different specialties. There was a greater delay (54 months) in establishing the diagnosis in those with atypical coeliac disease (p < 0.001). No difference was observed in the mode of delivery or duration of breast feeding, but significant difference was observed between gestational age at birth (p < 0.001). Most cases showed stunted growth and underweight. Irritability, anaemia, rickets, dermatitis herpetiformis, alopecia and intussusception were other common predictors of atypical coeliac disease. Because of a myriad spectrum of non-gastrointestinal symptoms, at any age with diverse presentation, a high index of suspicion is therefore required.
Collapse
Affiliation(s)
- Avinash Lomash
- PhD Scholar, Genetic Division, Department of Pediatrics, Maulana Azad Medical College, Lok Nayak Hospital, New Delhi, India
| | - Abhinaya Venkatakrishnan
- Senior Resident, Department of Pediatrics, Maulana Azad Medical College, Lok Nayak Hospital, New Delhi, India
| | - Meenakshi Bothra
- Assistant Professor, Department of Pediatrics, Maulana Azad Medical College, Lok Nayak Hospital, New Delhi, India
| | - Bhavna Dhingra
- Assistant Professor, Department of Pediatrics, All India Institute of Medical Sciences, Bhopal, India
| | - Praveen Kumar
- Director Professor, Department of Pediatrics, Lady Hardinge Medical College, Sucheta Kriplani Hospital, New Delhi, India
| | - Seema Kapoor
- Director Professor, Genetic division, Department of Pediatrics, Maulana Azad Medical College, Lok Nayak Hospital, New Delhi, India
| |
Collapse
|
25
|
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: 153] [Impact Index Per Article: 51.0] [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
|
26
|
Reduced peanut sensitization with maternal peanut consumption and early peanut introduction while breastfeeding. J Dev Orig Health Dis 2020; 12:811-818. [PMID: 33292902 DOI: 10.1017/s2040174420001129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
New guidelines for peanut allergy prevention in high-risk infants recommend introducing peanut during infancy but do not address breastfeeding or maternal peanut consumption. We assessed the independent and combined association of these factors with peanut sensitization in the general population CHILD birth cohort (N = 2759 mother-child dyads). Mothers reported peanut consumption during pregnancy, timing of first infant peanut consumption, and length of breastfeeding duration. Child peanut sensitization was determined by skin prick testing at 1, 3, and 5 years. Overall, 69% of mothers regularly consumed peanuts and 36% of infants were fed peanut in the first year (20% while breastfeeding and 16% after breastfeeding cessation). Infants who were introduced to peanut early (before 1 year) after breastfeeding cessation had a 66% reduced risk of sensitization at 5 years compared to those who were not (1.9% vs. 5.8% sensitization; aOR 0.34, 95% CI 0.14-0.68). This risk was further reduced if mothers introduced peanut early while breastfeeding and regularly consumed peanut themselves (0.3% sensitization; aOR 0.07, 0.01-0.25). In longitudinal analyses, these associations were driven by a higher odds of outgrowing early sensitization and a lower odds of late-onset sensitization. There was no apparent benefit (or harm) from maternal peanut consumption without breastfeeding. Taken together, these results suggest the combination of maternal peanut consumption and breastfeeding at the time of peanut introduction during infancy may help to decrease the risk of peanut sensitization. Mechanistic and clinical intervention studies are needed to confirm and understand this "triple exposure" hypothesis.
Collapse
|
27
|
Khan MR, Faubion WA, Dyer R, Singh R, Larson JJ, Absah I. Role of Lactulose Rhamnose Permeability Test in Assessing Small Bowel Mucosal Damage in Children with Celiac Disease. Glob Pediatr Health 2020; 7:2333794X20969278. [PMID: 33241082 PMCID: PMC7672748 DOI: 10.1177/2333794x20969278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/07/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Increased gut permeability has been suggested in patients with celiac disease (CD). We aimed to compare gut permeability in children using the lactulose/rhamnose permeability test. We prospectively recruited 55 children into 3 groups; 27 in group 1 (children with newly diagnosed celiac disease, 12 in group 2 (siblings of children with celiac disease) and 16 in group 3 (control group). The median age of participants was 11 years 2 months in group 1, 9 years 5 months in group 2 and 10 years 3 months in group 3. Standardized median delta rhamnose was lower in CD group as compared to control group (147.5 vs 3153.1, P = 0.040). The low median rhamnose absorption in children with celiac disease as compared to other groups suggests that this test can differentiate between damaged and healthy mucosa, hence can it potentially can be used as a noninvasive test of mucosal healing in children with celiac disease.
Collapse
Affiliation(s)
- Muhammad Rehan Khan
- University of Illinois College of Medicine at Peoria; Children’s Hospital of Illinois, Peroia, Illinois, USA
| | | | | | | | | | | |
Collapse
|
28
|
Martín-Masot R, Diaz-Castro J, Moreno-Fernandez J, Navas-López VM, Nestares T. The Role of Early Programming and Early Nutrition on the Development and Progression of Celiac Disease: A Review. Nutrients 2020; 12:nu12113427. [PMID: 33171615 PMCID: PMC7695164 DOI: 10.3390/nu12113427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 01/15/2023] Open
Abstract
Experimental and epidemiological evidence has shown that modifications of the intrauterine environment can have deleterious consequences for individuals, expressed as an increased risk of suffering non-communicable pathologies in adult life, which is known as the hypothesis of the early origin of diseases or fetal programming. On the other hand, changes in gene expression patterns through epigenetic modifications can be the basis for long-term maintenance of the effects of fetal programming. In this sense, epigenetics comprises the study of intrauterine disturbances, which develop diseases in the adult, including celiac disease (CD). In addition, early feeding practices could influence the risk of CD development, such as breastfeeding timing and duration and age of gluten introduction in the diet. Gluten acts as a trigger for CD in genetically predisposed subjects, although approximately 30% of the world population has HLA DQ2 or DQ8, the prevalence of the disease is only 1–3%. It is not known what factors act to modify the risk of disease in genetically at-risk subjects. Taking into account all these considerations, the aim of the current review is to elucidate the role of early programming and the effect of early nutrition on the development and progression of CD. It is logical that attention has been paid to gluten as a key element in preventing the disease. However, there is no strong evidence in favor of the protective factor of breastfeeding, timing of introduction of gluten during lactation, and the development of CD. Diet, genetic risk, microbiota, and environmental interaction are possible triggers of the change in tolerance to an immune response to gluten, but large-scale cohort studies are needed. Emerging scientific concepts, such as epigenetics, may help us establish the role of these factors.
Collapse
Affiliation(s)
- Rafael Martín-Masot
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (R.M.-M.); (V.M.N.-L.)
| | - Javier Diaz-Castro
- Department of Physiology and Institute of Nutrition and Food Technology “José MataixVerdú”, Biomedical Research Centre, University of Granada, 18010 Granada, Spain; (J.D.-C.); (J.M.-F.)
| | - Jorge Moreno-Fernandez
- Department of Physiology and Institute of Nutrition and Food Technology “José MataixVerdú”, Biomedical Research Centre, University of Granada, 18010 Granada, Spain; (J.D.-C.); (J.M.-F.)
| | - Víctor Manuel Navas-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (R.M.-M.); (V.M.N.-L.)
| | - Teresa Nestares
- Department of Physiology and Institute of Nutrition and Food Technology “José MataixVerdú”, Biomedical Research Centre, University of Granada, 18010 Granada, Spain; (J.D.-C.); (J.M.-F.)
- Correspondence: ; Tel.: +34-69-698-9989
| |
Collapse
|
29
|
Llorente Pelayo S, Palacios Sánchez M, Docio Pérez P, Gutiérrez Buendía D, Peña Sainz-Pardo E, Vega Santa-Cruz B, García Calatayud S. [Infections in early life as risk factor for coeliac disease]. An Pediatr (Barc) 2020; 94:293-300. [PMID: 33011112 DOI: 10.1016/j.anpedi.2020.06.022] [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: 04/30/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Among the environmental factors that can affect the pathological response to gluten in coeliac disease (CD), the factors that influence the immune response, such as infections and use of antibiotics, are proposed. Our objective is to determine the relationship between infections in early life and the risk of CD. PATIENTS AND METHODS A retrospective case-control study, including patients aged 0-16 years with a diagnosis of CD was performed between the years 2014-2018. An analysis was made of documented infections in the first 6 months of life, types of infection (respiratory, gastrointestinal, urinary, others), microorganisms involved, and antibiotic therapy used. RESULTS A total of 93 coeliac patients, 93 controls, and 237 infectious episodes were registered. Documented infections affected 67.7% of coeliac patients and 50.5% of controls (P = .017), with a mean of 1.49 ± 1.53 episodes in the coeliac group and 1.05 ± 1.5 in the controls (P = .016). Documented infections in the first 6 months of life doubles the risk of developing CD (OR 2.05; 95% CI; 1.13-3.73), with this risk being higher for respiratory infections, which multiply the risk by 2.3 (OR 2.30, 95% CI; 1.28-4.14). Also, having 3 or more respiratory infections in the first 6 months of life multiplied the risk by 2.8 (OR 2.79, 95% CI; 1.03-7.54). No differences were found related to the types of involved microorganism or regarding the use of antibiotics. CONCLUSIONS Infections in the first 6 months of life increase the risk of developing CD, especially for respiratory infections and, to a greater extent, if 3 or more episodes occur. The use of antibiotics in this period of life has not been related to an increased risk of CD.
Collapse
Affiliation(s)
| | - Mirian Palacios Sánchez
- Unidad de Gastroenterología Infantil. Servicio de Pediatría. Hospital Universitario Marqués de Valdecilla, Santander, España
| | - Pablo Docio Pérez
- Servicio de Pediatría. Hospital Universitario Marqués de Valdecilla, Santander, España
| | | | | | | | - Salvador García Calatayud
- Unidad de Gastroenterología Infantil. Servicio de Pediatría. Hospital Universitario Marqués de Valdecilla, Santander, España
| |
Collapse
|
30
|
Serena G, D'Avino P, Fasano A. Celiac Disease and Non-celiac Wheat Sensitivity: State of Art of Non-dietary Therapies. Front Nutr 2020; 7:152. [PMID: 33015123 PMCID: PMC7506149 DOI: 10.3389/fnut.2020.00152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Gluten related disorders (GRD), which include celiac disease, non-celiac wheat sensitivity and wheat allergy are heterogeneous conditions triggered by ingestion of gluten-containing grains. Together, their prevalence is estimated to be ~5% in the general population, however, in the last years the number of diagnoses has been rapidly increasing. To this day, the gold standard treatment for these disorders is the complete removal of gluten-containing grains from the diet. Although this therapy results effective in the majority of patients, up to 30% of individuals affected by GRD continue to present persistent symptoms. In addition, gluten-free diet has been shown to have poor nutritional quality and to cause a socio-economic burden in patients' quality of life. In order to respond to these issues, the scientific community has been focusing on finding additional and adjuvant non-dietary therapies. In this review, we focus on two main gluten related disorders, celiac disease and non-celiac wheat sensitivity. We delineate the actual knowledge about potential treatments and their relative efficacy in pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Paolo D'Avino
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States.,Vita-Salute San Raffaele University, Milan, Italy
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States.,European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| |
Collapse
|
31
|
Pecora F, Persico F, Gismondi P, Fornaroli F, Iuliano S, de'Angelis GL, Esposito S. Gut Microbiota in Celiac Disease: Is There Any Role for Probiotics? Front Immunol 2020; 11:957. [PMID: 32499787 PMCID: PMC7243837 DOI: 10.3389/fimmu.2020.00957] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated disorder initiated by the ingestion of gluten in genetically predisposed individuals. Recent data shows that changes in the gut microbiome composition and function are linked with chronic inflammatory diseases; this might also be the case for CD. The main aim of this manuscript is to discuss our present knowledge of the relationships between gut microbiota alterations and CD and to understand if there is any role for probiotics in CD therapy. PubMed was used to search for all of the studies published from November 2009 to November 2019 using key words such as “Celiac Disease” and “Microbiota” (306 articles), “Celiac Disease” and “Gastrointestinal Microbiome” (139), and “Probiotics” and “Celiac Disease” (97 articles). The search was limited to articles published in English that provided evidence-based data. Literature analysis showed that the gut microbiota has a well-established role in gluten metabolism, in modulating the immune response and in regulating the permeability of the intestinal barrier. Promising studies suggest a possible role of probiotics in treating and/or preventing CD. Nevertheless, human trials on the subject are still scarce and lack homogeneity. A possible role was documented for probiotics in improving CD-related symptoms, modulating the peripheral immune response and altering the fecal microbiota, although the results were not consistent in all of the studies. No evidence was found that probiotic administration might prevent CD onset. Knowledge of the role of intestinal bacteria in the development of CD opens new possibilities for its treatment through probiotic administration, even though further studies are needed to better clarify whether probiotics can help treat or prevent the disease and to define which probiotics to use, at what dose and for how long.
Collapse
Affiliation(s)
- Francesco Pecora
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | - Federica Persico
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | - Pierpacifico Gismondi
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | - Fabiola Fornaroli
- Unit of Gastroenterology and Digestive Endoscopy, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia Iuliano
- Unit of Gastroenterology and Digestive Endoscopy, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gian Luigi de'Angelis
- Unit of Gastroenterology and Digestive Endoscopy, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| |
Collapse
|
32
|
Abstract
Gluten-related disorders, including celiac disease, wheat allergy, and nonceliac gluten sensitivity (NCGS), are increasingly reported worldwide. Celiac disease is caused by an immune-mediated reaction to ingested gluten in genetically susceptible persons. NCGS is largely a diagnosis of exclusion when other causes of symptoms have been ruled out. All patients with celiac disease should be referred to a registered dietitian nutritionist with expertise in celiac disease and a gastroenterologist who specializes in celiac disease and malabsorptive disorders, and they should remain on a strict gluten-free diet indefinitely. This article provides an overview of gluten- and wheat-related disorders.
Collapse
Affiliation(s)
- Joshua Elliott Rubin
- University of California, San Diego School of Medicine, La Jolla, California (J.E.R., S.E.C.)
| | - Sheila E Crowe
- University of California, San Diego School of Medicine, La Jolla, California (J.E.R., S.E.C.)
| |
Collapse
|
33
|
Mirza AH, Kaur S, Nielsen LB, Størling J, Yarani R, Roursgaard M, Mathiesen ER, Damm P, Svare J, Mortensen HB, Pociot F. Breast Milk-Derived Extracellular Vesicles Enriched in Exosomes From Mothers With Type 1 Diabetes Contain Aberrant Levels of microRNAs. Front Immunol 2019; 10:2543. [PMID: 31708933 PMCID: PMC6823203 DOI: 10.3389/fimmu.2019.02543] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022] Open
Abstract
The breast milk plays a crucial role in shaping the initial intestinal microbiota and mucosal immunity of the infant. Interestingly, breastfeeding has proven to be protective against the early onset of immune-mediated diseases including type 1 diabetes. Studies have shown that exosomes from human breast milk are enriched in immune-modulating miRNAs suggesting that exosomal miRNAs (exomiRs) transferred to the infant could play a critical role in the development of the infant's immune system. We extracted exomiRs from breast milk of 52 lactating mothers (26 mothers with type 1 diabetes and 26 healthy mothers), to identify any differences in the exomiR content between the two groups. Small RNA-sequencing was performed to identify known and novel miRNAs in both groups. A total of 631 exomiRs were detected by small RNA sequencing including immune-related miRNAs such as hsa-let-7c, hsa-miR-21, hsa-miR-34a, hsa-miR-146b, and hsa-miR-200b. In addition, ~200 novel miRNAs were identified in both type 1 diabetes and control samples. Among the known miRNAs, nine exomiR's were found differentially expressed in mothers with type 1 diabetes compared to healthy mothers. The highly up-regulated miRNAs, hsa-miR-4497, and hsa-miR-3178, increased lipopolysaccharide-induced expression and secretion of tumor necrosis factor α (TNFα) in human monocytes. The up-regulated miRNA target genes were significantly enriched for longevity-regulating pathways and FoxO signaling. Our findings suggest a role of breast milk-derived exomiRs in modulating the infant's immune system.
Collapse
Affiliation(s)
- Aashiq H Mirza
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Simranjeet Kaur
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lotte B Nielsen
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Joachim Størling
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Reza Yarani
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Martin Roursgaard
- Faculty of Health Sciences, Institute of Public Health, CSS, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth R Mathiesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Damm
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Pregnant Women With Diabetes, Rigshospitalet, Copenhagen, Denmark.,Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Jens Svare
- Department of Obstetrics, Herlev Hospital, Herlev, Denmark
| | - Henrik B Mortensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Flemming Pociot
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
34
|
The Gut Microbiota in Celiac Disease and probiotics. Nutrients 2019; 11:nu11102375. [PMID: 31590358 PMCID: PMC6836185 DOI: 10.3390/nu11102375] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CeD) is an immune-mediated enteropathy, and unique in that the specific trigger is known: gluten. The current mainstay of therapy is a gluten-free diet (GFD). As novel therapies are being developed, complementary strategies are also being studied, such as modulation of the gut microbiome. The gut microbiota is involved in the initiation and perpetuation of intestinal inflammation in several chronic diseases. Intestinal dysbiosis has been reported in CeD patients, untreated or treated with GFD, compared to healthy subjects. Several studies have identified differential bacterial populations associated with CeD patients and healthy subjects. However, it is still not clear if intestinal dysbiosis is the cause or effect of CeD. Probiotics have also been considered as a strategy to modulate the gut microbiome to an anti-inflammatory state. However, there is a paucity of data to support their use in treating CeD. Further studies are needed with therapeutic microbial formulations combined with human trials on the use of probiotics to treat CeD by restoring the gut microbiome to an anti-inflammatory state.
Collapse
|
35
|
Jones CL, Culpin I, Evans J, Pearson RM. Relative effects of breastfeeding intention and practice on maternal responsiveness. Infant Ment Health J 2019; 41:82-93. [PMID: 31553493 PMCID: PMC7028075 DOI: 10.1002/imhj.21832] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Our objective was to examine the differential effects of antenatal breastfeeding intention (BI) and breastfeeding practice (BP) on maternal postnatal responsiveness. We conducted a secondary analysis of longitudinal data from a subsample of 962 mother-infant dyads from a U.K.-based birth cohort study the Avon Longitudinal Study of Parents and Children. Exposures were BI and BPs measured at 32 weeks of gestation and 18 months' postpartum. The outcome was maternal responsiveness assessed at 12 months' postpartum. We used logistic regression analyses unadjusted and adjusted for confounders. Intention to breastfeed was associated with increased odds of postnatal maternal responsiveness independent of BP, adjusted odds ratio (OR) = 2.34, 95% CI [1.42, 3.86]. There was no evidence that BP was an independent predictor of maternal responsiveness, OR = 0.93, 95% CI [0.55, 1.57]. Life-course epidemiology analyses demonstrated that maternal responsiveness is most positive when both BI and BP are present. This is the first population-based study to provide evidence that BI during pregnancy is more strongly associated with maternal postnatal responsiveness than is BP. Further research is needed to understand the determinants of BI in pregnancy and its relationships with maternal responsiveness.
Collapse
Affiliation(s)
- Catherine L Jones
- Department of Psychology, School of Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Iryna Culpin
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jonathan Evans
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Rebecca M Pearson
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
36
|
Castenmiller J, de Henauw S, Hirsch-Ernst KI, Kearney J, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bresson JL, Fewtrell M, Kersting M, Przyrembel H, Dumas C, Titz A, Turck D. Appropriate age range for introduction of complementary feeding into an infant's diet. EFSA J 2019; 17:e05780. [PMID: 32626427 PMCID: PMC7009265 DOI: 10.2903/j.efsa.2019.5780] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Following a request from the European Commission, the Panel on Nutrition, Novel Foods and Food Allergens (NDA) revised its 2009 Opinion on the appropriate age for introduction of complementary feeding of infants. This age has been evaluated considering the effects on health outcomes, nutritional aspects and infant development, and depends on the individual's characteristics and development. As long as foods have an age-appropriate texture, are nutritionally appropriate and prepared following good hygiene practices, there is no convincing evidence that at any age investigated in the included studies (< 1 to < 6 months), the introduction of complementary foods (CFs) is associated with adverse health effects or benefits (except for infants at risk of iron depletion). For nutritional reasons, the majority of infants need CFs from around 6 months of age. Infants at risk of iron depletion (exclusively breastfed infants born to mothers with low iron status, or with early umbilical cord clamping (< 1 min after birth), or born preterm, or born small-for-gestational age or with high growth velocity) may benefit from earlier introduction of CFs that are a source of iron. The earliest developmental skills relevant for consuming pureed CFs can be observed between 3 and 4 months of age. Skills for consuming finger foods can be observed in some infants at 4 months, but more commonly at 5-7 months. The fact that an infant may be ready from a neurodevelopmental perspective to progress to a more diversified diet before 6 months of age does not imply that there is a need to introduce CFs. There is no reason to postpone the introduction of potentially allergenic foods (egg, cereals, fish and peanut) to a later age than that of other CFs as far as the risk of developing atopic diseases is concerned. Regarding the risk of coeliac disease, gluten can be introduced with other CFs.
Collapse
|
37
|
Popp A, Mäki M. Changing Pattern of Childhood Celiac Disease Epidemiology: Contributing Factors. Front Pediatr 2019; 7:357. [PMID: 31555624 PMCID: PMC6727179 DOI: 10.3389/fped.2019.00357] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/15/2019] [Indexed: 12/14/2022] Open
Abstract
Up until the 1960s and 1970s, diarrhea, malabsorption syndrome, and failure to thrive were the presenting symptoms and signs of celiac disease (CD) in young infants; however this disease was also at the same time reported to be disappearing. Indeed, clinical childhood CD was seen to transform into a milder form, resulting in an upward shift in age at diagnosis during the 1970s (and years later for many countries). This changing pattern of CD presentation then altered the epidemiology of the disease, with major differences between and within countries observed. An awareness of the changing clinical nature of CD and use of case-finding tools to detect even clinically silent CD became an important factor in this changing epidemiology. Countries report both low and high prevalence but it seems to be on the increase resulting in a population-based level of 1-2%. This paper discusses the potential causes and environmental factors behind these observed clinical changes, identifying new clues from different studies published at the time this transformation took place. For instance, it was found that breastfeeding postponed the diagnosis of the disease but did not altogether prevent it. Moreover, gluten introduction at a young age, specifically at the mean age of 2 months, seemed to also have a clear impact in inducing malabsorption syndrome and failure to thrive in young infants in addition to other factors such as gluten intake volume and type of cereal present in the weaning food. Further, the impact of cow's milk and its high osmolarity might have played an important role; humanized milk formulas were not yet invented. Future epidemiological studies on the contributing environmental factors to the shift in CD presentation are thus recommended for countries in which these changing clinical features are still being observed.
Collapse
Affiliation(s)
- Alina Popp
- Faculty of Medicine and Health Technology, Tampere Center of Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
- National Institute for Mother and Child Health “Alessandrescu-Rusescu”, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Markku Mäki
- Faculty of Medicine and Health Technology, Tampere Center of Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
38
|
Affiliation(s)
- Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts
- Center for Celiac Research and Treatment, Mucosal Immunology and Biology Research Center, Boston, Massachusetts
- Celiac Research Program, Harvard Medical School, Boston, Massachusetts
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts
- Center for Celiac Research and Treatment, Mucosal Immunology and Biology Research Center, Boston, Massachusetts
- Celiac Research Program, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Andrén Aronsson C, Lee HS, Hård af Segerstad EM, Uusitalo U, Yang J, Koletzko S, Liu E, Kurppa K, Bingley PJ, Toppari J, Ziegler AG, She JX, Hagopian WA, Rewers M, Akolkar B, Krischer JP, Virtanen SM, Norris JM, Agardh D. Association of Gluten Intake During the First 5 Years of Life With Incidence of Celiac Disease Autoimmunity and Celiac Disease Among Children at Increased Risk. JAMA 2019; 322:514-523. [PMID: 31408136 PMCID: PMC6692672 DOI: 10.1001/jama.2019.10329] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPORTANCE High gluten intake during childhood may confer risk of celiac disease. OBJECTIVES To investigate if the amount of gluten intake is associated with celiac disease autoimmunity and celiac disease in genetically at-risk children. DESIGN, SETTING, AND PARTICIPANTS The participants in The Environmental Determinants of Diabetes in the Young (TEDDY), a prospective observational birth cohort study designed to identify environmental triggers of type 1 diabetes and celiac disease, were followed up at 6 clinical centers in Finland, Germany, Sweden, and the United States. Between 2004 and 2010, 8676 newborns carrying HLA antigen genotypes associated with type 1 diabetes and celiac disease were enrolled. Screening for celiac disease with tissue transglutaminase autoantibodies was performed annually in 6757 children from the age of 2 years. Data on gluten intake were available in 6605 children (98%) by September 30, 2017. EXPOSURES Gluten intake was estimated from 3-day food records collected at ages 6, 9, and 12 months and biannually thereafter until the age of 5 years. MAIN OUTCOMES AND MEASURES The primary outcome was celiac disease autoimmunity, defined as positive tissue transglutaminase autoantibodies found in 2 consecutive serum samples. The secondary outcome was celiac disease confirmed by intestinal biopsy or persistently high tissue transglutaminase autoantibody levels. RESULTS Of the 6605 children (49% females; median follow-up: 9.0 years [interquartile range, 8.0-10.0 years]), 1216 (18%) developed celiac disease autoimmunity and 447 (7%) developed celiac disease. The incidence for both outcomes peaked at the age of 2 to 3 years. Daily gluten intake was associated with higher risk of celiac disease autoimmunity for every 1-g/d increase in gluten consumption (hazard ratio [HR], 1.30 [95% CI, 1.22-1.38]; absolute risk by the age of 3 years if the reference amount of gluten was consumed, 28.1%; absolute risk if gluten intake was 1-g/d higher than the reference amount, 34.2%; absolute risk difference, 6.1% [95% CI, 4.5%-7.7%]). Daily gluten intake was associated with higher risk of celiac disease for every 1-g/d increase in gluten consumption (HR, 1.50 [95% CI, 1.35-1.66]; absolute risk by age of 3 years if the reference amount of gluten was consumed, 20.7%; absolute risk if gluten intake was 1-g/d higher than the reference amount, 27.9%; absolute risk difference, 7.2% [95% CI, 6.1%-8.3%]). CONCLUSIONS AND RELEVANCE Higher gluten intake during the first 5 years of life was associated with increased risk of celiac disease autoimmunity and celiac disease among genetically predisposed children.
Collapse
Affiliation(s)
| | - Hye-Seung Lee
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa
| | | | - Ulla Uusitalo
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa
| | - Jimin Yang
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa
| | - Sibylle Koletzko
- Dr von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
- University of Warmia and Mazuri, Olsztyn, Poland
| | - Edwin Liu
- Digestive Health Institute, University of Colorado Denver, Children’s Hospital Colorado, Denver
| | - Kalle Kurppa
- Tampere Centre for Child Health Research, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Polly J. Bingley
- School of Clinical Sciences, University of Bristol, Bristol, England
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Anette G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Klinikum rechts der Isar, Technische Universität München, and Forschergruppe Diabetes eV, Neuherberg, Germany
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia
| | | | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Jeffrey P. Krischer
- Health Informatics Institute, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa
| | - Suvi M. Virtanen
- National Institute for Health and Welfare, Department of Public Health Solutions, Helsinki, Finland
- Faculty of Social Sciences/Health Sciences, University of Tampere, Tampere, Finland
- Research Center for Child Health, Tampere University, University Hospital, Science Center of Pirkanmaa Hospital District, Tampere, Finland
| | - Jill M. Norris
- Colorado School of Public Health, Department of Epidemiology, University of Colorado, Aurora
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
40
|
Gluten Intake in Early Childhood and Risk of Celiac Disease in Childhood: A Nationwide Cohort Study. Am J Gastroenterol 2019; 114:1299-1306. [PMID: 31343439 DOI: 10.14309/ajg.0000000000000331] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Celiac disease (CD) may occur in genetically predisposed individuals exposed to gluten, but it is unclear whether the amount of gluten influences the risk of disease. We aimed at determining whether the amount of gluten intake at age 18 months predicted later risk of CD. METHODS In an observational nationwide cohort study, the Norwegian Mother and Child Cohort Study (MoBa), we included 67,608 children born during 2000-2009 and followed up for a mean of 11.5 years (range 7.5-15.5) after exclusions for missing data. Information regarding CD diagnosis was obtained from the Norwegian Patient Register 2008-2016 and from parental questionnaires at child age 7 and 8 years. We estimated gluten intake at age 18 months from a prospectively collected parental questionnaire. RESULTS CD was diagnosed in 738 children (1.1%, 62% girls). The mean estimated amount of gluten in the diet at 18 months was 8.8 g/d (SD 3.6). The adjusted relative risk of CD was 1.10 (95% confidence interval 1.03-1.18) per SD increase in daily gluten amount at age 18 months. Children in the upper quartile of gluten intake compared with the lower quartile had an increased risk of CD (adjusted relative risk 1.29, 95% confidence interval 1.06-1.58). The association with gluten amount was independent of the age at introduction of gluten. Gluten introduction ≥6 months was also an independent risk factor for CD. DISCUSSION In this nationwide study, increased gluten intake at 18 months was associated with a modestly increased risk of CD later in childhood.
Collapse
|
41
|
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
|
42
|
Abstract
The prevalence of celiac disease (CeD) has increased in the last decades, suggesting a role for environmental factors in addition to gluten. Several cohort studies have shown that different gastrointestinal infections increase CeD risk. However, the mechanisms by which microbes participate in CeD have remained elusive. Recently, with the use of animal models, both viral and bacterial opportunistic pathogens were shown to induce immune activation relevant for CeD. The hypothesis that viral and/or bacterial infections can contribute to immune activation and breakdown of tolerance toward gluten in genetically susceptible individuals is therefore reinforced. Here, we discuss the evidence regarding the role of microbes in promoting CeD and the specific pathways triggered by microbes that could participate in CeD pathogenesis. Understanding these pathways will allow us to develop optimal microbiota-modulating strategies to help prevent CeD.
Collapse
Affiliation(s)
- Alberto Caminero
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Elena F. Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
43
|
Wheat Allergy in Children: A Comprehensive Update. ACTA ACUST UNITED AC 2019; 55:medicina55070400. [PMID: 31340608 PMCID: PMC6681225 DOI: 10.3390/medicina55070400] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/14/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022]
Abstract
Gluten-related disorders are very common in pediatric patients. Wheat allergy is triggered by an immunoglobulin E (IgE)-dependent mechanism; its prevalence varies according to the age and region, and in Europe has been estimated to be lower than 1%. Many studies investigated the potential role of several external factors that can influence the risk to developing wheat allergy, but results are still inconclusive. It can be responsible for several clinical manifestations depending on the route of allergen exposure: food-dependent exercise-induced anaphylaxis (FDEIA), occupational rhinitis or asthma (also known as baker’s asthma), and contact urticaria. The prognosis of IgE-mediated wheat allergy in children is generally favorable, with the majority of children becoming tolerant by school age. Patients who experienced an anaphylactic reaction prior to 3 years of age and patients with higher level of wheat- or ω-5 gliadin-specific IgE antibodies seem to be at higher risk of persistent wheat allergy. The current management of patients is dietary avoidance. Nowadays, oral immunotherapy has been proposed for wheat allergy with promising results, even if further studies are necessary to establish the best protocol in order to promote tolerance in wheat-allergic children.
Collapse
|
44
|
Güngör D, Nadaud P, Dreibelbis C, LaPergola CC, Wong YP, Terry N, Abrams SA, Beker L, Jacobovits T, Järvinen KM, Nommsen-Rivers LA, O'Brien KO, Oken E, Pérez-Escamilla R, Ziegler EE, Spahn JM. Infant milk-feeding practices and diagnosed celiac disease and inflammatory bowel disease in offspring: a systematic review. Am J Clin Nutr 2019; 109:838S-851S. [PMID: 30982875 PMCID: PMC6500925 DOI: 10.1093/ajcn/nqy371] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND During the Pregnancy and Birth to 24 Months Project, the USDA and US Department of Health and Human Services initiated an evidence review on diet and health in these populations. OBJECTIVE The aim of these systematic reviews was to examine the relationships of never versus ever feeding human milk, shorter versus longer durations of any and exclusive human milk feeding, and feeding a lower versus a higher intensity of human milk to mixed-fed infants with diagnosed celiac disease and inflammatory bowel disease (IBD). METHODS The Nutrition Evidence Systematic Review team (formerly called the Nutrition Evidence Library) conducted systematic reviews with external experts. We searched CINAHL, Cochrane, Embase, and PubMed for articles published January, 1980 to March, 2016, dual-screened the results using predetermined criteria, extracted data from and assessed risk of bias for each included study, qualitatively synthesized the evidence, developed conclusion statements, and graded the strength of the evidence. RESULTS We included 9 celiac disease and 17 IBD articles. Limited case-control evidence suggests never versus ever being fed human milk is associated with higher risk of celiac disease, but concerns about reverse causality precluded a conclusion about the relationship of shorter versus longer durations of any human milk feeding with celiac disease. Evidence examining never versus ever feeding human milk and IBD was inconclusive, and limited, but consistent, case-control evidence suggests that, among infants fed human milk, shorter versus longer durations of any human milk feeding are associated with higher risk of IBD. For both outcomes, evidence examining the duration of exclusive human milk feeding was scant and no articles examined the intensity of human milk fed to mixed-fed infants. CONCLUSION Limited case-control evidence suggests that feeding human milk for short durations or not at all associates with higher risk of diagnosed IBD and celiac disease, respectively. The small number of studies and concern about reverse causality and recall bias prevent stronger conclusions.
Collapse
Affiliation(s)
| | | | | | | | | | - Nancy Terry
- National Institutes of Health Library, Bethesda, MD
| | - Steve A Abrams
- Dell Medical School at the University of Texas, Austin, TX
| | - Leila Beker
- US Food and Drug Administration, contractor, College Park, MD
| | | | | | | | | | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Rafael Pérez-Escamilla
- Department of Social and Behavioral Sciences, Yale School of Public Health, New Haven, CT
| | | | | |
Collapse
|
45
|
Abstract
Coeliac disease (CD) is an immune-mediated disorder triggered by the ingestion of gluten in genetically susceptible individuals. However, only a small proportion of subjects harbouring CD-related genetic risk develop the disease. Among the environmental factors that may influence CD risk, pre- and perinatal factors, delivery methods, parental lifestyle, infant feeding practices, seasonality, dietary factors, drug use, childhood infections and variability in gut microbiota are those most widely studied regarding the risk to develop CD. Although for many of these external factors the exact mechanism of action is unknown, most of them are thought to act by disrupting the intestinal barrier, facilitating contact between potential antigens and the immune system effector cells. Management of CD is relatively easy in patients with a definite diagnosis and requires a strict, lifelong, gluten-free diet. Better knowledge of environmental exposures apart from gluten can facilitate understanding of the pathogenesis of the disorder and the wide heterogeneity of its clinical spectrum. The purpose of this review is to discuss current knowledge on environmental CD risk factors, as well as possible interaction between them, on the grounds of the reliable scientific evidence available. Key messages The risk of developing CD is influenced not only by gluten ingestion but also by a number of environmental factors including childhood infections and variability in gut microbiota, pre- and perinatal factors, infant feeding practices, delivery methods, parental lifestyle, seasonality, dietary factors and drug use, acting mainly by disrupting intestinal permeability. Better knowledge of exposure to these factors can facilitate their identification, and subsequent elimination, in the individual patient.
Collapse
Affiliation(s)
- Giovanni Mario Pes
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy
| | - Stefano Bibbò
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy
| | - Maria Pina Dore
- a Department of Medical , Surgical and Experimental Sciences, University of Sassari , Sassari , Italy.,b Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
46
|
Crespo Escobar P, Castillejo G, Martínez-Ojinaga E, Donat E, Polanco I, Mearin ML, Ribes-Koninckx C. Ten years of follow-up of the Spanish cohort of the European PreventCD study: the lessons learned. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2018; 110:493-499. [PMID: 29699403 DOI: 10.17235/reed.2018.5324/2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIM to evaluate the influence of gluten consumption on celiac disease development and to describe its natural history in the Spanish cohort of the European PreventCD study. METHODS prospective multi-center double blind study of 225 children that were followed up from birth. All cases were HLA-DQ2/HLA-DQ8 positive with a 1st degree relative with celiac disease and were followed up in three centers from Madrid, Reus and Valencia. Gluten intake was determined between four and ten months according to the protocol. Gluten intake was ad libitum between eleven and 36 months and was prospectively quantified by means of dietary records. Clinical visits and specific antibody analysis for celiac disease were performed periodically. RESULTS twenty-six cases were diagnosed, all had a positive biopsy and serology; 21 had gastrointestinal symptoms and five were asymptomatic. In addition, 2,565 food records were analyzed and statistically significant differences (p < 0.001) were found with regard to gluten consumption among the three centers, although not between celiac and non-celiac children (p = 0.025). The HLA-DQ2.5/DQ2.5 and DQ2.5/DQ2.2 genotypes had a relative risk of 4.7 (95% CI: 0.80-27.55; p = 0.08), which was higher than for the rest of genotypes. Female gender also had a relative risk that was five times higher than that for males. CONCLUSIONS the amount of gluten intake between 11 and 36 months or the duration of breast feeding were not risk factors for the development of CD in the Spanish population. The HLA genotype and gender were the most relevant associated factors. In this at-risk group, the disease presented before two years of age in the majority of the cases with a weak clinical expression.
Collapse
Affiliation(s)
- Paula Crespo Escobar
- Unidad de Enfermedad Celiaca e Inmunopatología Dig, Instituto de Investigación Sanitaria la Fe, España
| | - Gemma Castillejo
- Hospital Universitari Sant Joan, Reus y Universitat Rovira i Virgili, España
| | | | - Ester Donat
- Gastroenterología y Hepatología Pediátrica,, Hospital Universitario y Politécnico La Fe,, España,
| | | | | | - Carmen Ribes-Koninckx
- Gastroenterología y Hepatología Pediátrica,, Hospital Universitario y Politécnico La Fe,
| |
Collapse
|
47
|
Barroso M, Beth SA, Voortman T, Jaddoe VWV, van Zelm MC, Moll HA, Kiefte-de Jong JC. Dietary Patterns After the Weaning and Lactation Period Are Associated With Celiac Disease Autoimmunity in Children. Gastroenterology 2018; 154:2087-2096.e7. [PMID: 29481779 DOI: 10.1053/j.gastro.2018.02.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/03/2018] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS There have been many studies of associations between infant feeding practices and development of celiac disease during childhood, but few studies have focused on overall diets of young children after the weaning period. We aimed to examine the association between common dietary patterns in infants and the occurrence of celiac disease autoimmunity during childhood. METHODS We performed a prospective analysis of data from the Generation R Study that comprised 1997 children born from April 2002 through January 2006 in Rotterdam, the Netherlands. Food consumption around 1 year of age was assessed with a validated food-frequency questionnaire. Dietary data were examined using a priori (based on existing guidelines) and a posteriori (principal component analysis and reduced rank regression) dietary pattern analyses. Five dietary patterns were compared. Celiac disease autoimmunity, determined on the basis of serum concentration of transglutaminase-2 autoantibody (ie, TG2A) below or above 7 U/mL, was evaluated at 6 years. Associations between dietary pattern adherence scores and celiac disease autoimmunity were examined using multivariable logistic regression models. RESULTS Higher adherence to the a posteriori-derived prudent dietary pattern (high intake of vegetables, vegetable oils, pasta, and grains and low consumption of refined cereals and sweet beverages) at 1 year was significantly associated with lower odds of celiac disease autoimmunity at 6 years (odds ratio, 0.67; 95% confidence interval, 0.53-0.84). No significant associations were found for the 4 remaining dietary patterns. CONCLUSIONS In a prospective study of dietary patterns of young children in the Netherlands, we associated a dietary pattern characterized by high consumption of vegetables and grains and low consumption of refined cereals and sweet beverages, with lower odds of celiac disease autoimmunity. Early-life dietary patterns might therefore be involved in the development of celiac disease during childhood.
Collapse
Affiliation(s)
- Monica Barroso
- Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sytske A Beth
- Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Vincent W V Jaddoe
- Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Henriette A Moll
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jessica C Kiefte-de Jong
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Leiden University College, the Hague, the Netherlands.
| |
Collapse
|
48
|
Hård Af Segerstad EM, Lee HS, Andrén Aronsson C, Yang J, Uusitalo U, Sjöholm I, Rayner M, Kurppa K, Virtanen SM, Norris JM, Agardh D. Daily Intake of Milk Powder and Risk of Celiac Disease in Early Childhood: A Nested Case-Control Study. Nutrients 2018; 10:E550. [PMID: 29710789 PMCID: PMC5986430 DOI: 10.3390/nu10050550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 01/29/2023] Open
Abstract
Milk powder and gluten are common components in Swedish infants' diets. Whereas large intakes of gluten early in life increases the risk of celiac disease in genetically at-risk Swedish children, no study has yet evaluated if intake of milk powder by 2 years of age is associated with celiac disease. A 1-to-3 nested case-control study, comprised of 207 celiac disease children and 621 controls matched for sex, birth year, and HLA genotype, was performed on a birth cohort of HLA-DR3-DQ2 and/or DR4-DQ8-positive children. Subjects were screened annually for celiac disease using tissue transglutaminase autoantibodies (tTGA). Three-day food records estimated the mean intake of milk powder at ages 6 months, 9 months, 12 months, 18 months, and 24 months. Conditional logistic regression calculated odds ratios (OR) at last intake prior to seroconversion of tTGA positivity, and for each time-point respectively and adjusted for having a first-degree relative with celiac disease and gluten intake. Intake of milk powder prior to seroconversion of tTGA positivity was not associated with celiac disease (OR = 1.00; 95% CI = 0.99, 1.03; p = 0.763). In conclusion, intake of milk powder in early childhood is not associated with celiac disease in genetically susceptible children.
Collapse
Affiliation(s)
- Elin M Hård Af Segerstad
- The Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University, 202 05 Malmö, Sweden.
| | - Hye-Seung Lee
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, 33620 FL Tampa, USA.
| | - Carin Andrén Aronsson
- The Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University, 202 05 Malmö, Sweden.
| | - Jimin Yang
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, 33620 FL Tampa, USA.
| | - Ulla Uusitalo
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, 33620 FL Tampa, USA.
| | - Ingegerd Sjöholm
- Department of Food Technology, Engineering and Nutrition, Chemical Center, Lund University, 221 00 Lund, Sweden.
| | - Marilyn Rayner
- Department of Food Technology, Engineering and Nutrition, Chemical Center, Lund University, 221 00 Lund, Sweden.
| | - Kalle Kurppa
- Tampere Center for Child Health Research, University of Tampere and Tampere University Hospital, 33521 Tampere, Finland.
| | - Suvi M Virtanen
- Unit of Nutrition, National Institute for Health and Welfare, 00271 Helsinki, Finland; Faculty of Social Sciences, University of Tampere, Tampere Center for Child Health Research, University of Tampere and Tampere University Hospital and the Science Center of Pirkanmaa Hospital District Tampere, 33521 Tampere, Finland.
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, 80045 CO Aurora, USA.
| | - Daniel Agardh
- The Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University, 202 05 Malmö, Sweden.
| |
Collapse
|
49
|
Rintala A, Riikonen I, Toivonen A, Pietilä S, Munukka E, Pursiheimo JP, Elo LL, Arikoski P, Luopajärvi K, Schwab U, Uusitupa M, Heinonen S, Savilahti E, Eerola E, Ilonen J. Early fecal microbiota composition in children who later develop celiac disease and associated autoimmunity. Scand J Gastroenterol 2018; 53:403-409. [PMID: 29504486 DOI: 10.1080/00365521.2018.1444788] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Several studies have reported that the intestinal microbiota composition of celiac disease (CD) patients differs from healthy individuals. The possible role of gut microbiota in the pathogenesis of the disease is, however, not known. Here, we aimed to assess the possible differences in early fecal microbiota composition between children that later developed CD and healthy controls matched for age, sex and HLA risk genotype. MATERIALS AND METHODS We used 16S rRNA gene sequencing to examine the fecal microbiota of 27 children with high genetic risk of developing CD. Nine of these children developed the disease by the age of 4 years. Stool samples were collected at the age of 9 and 12 months, before any of the children had developed CD. The fecal microbiota composition of children who later developed the disease was compared with the microbiota of the children who did not have CD or associated autoantibodies at the age of 4 years. Delivery mode, early nutrition, and use of antibiotics were taken into account in the analyses. RESULTS No statistically significant differences in the fecal microbiota composition were found between children who later developed CD (n = 9) and the control children without disease or associated autoantibodies (n = 18). CONCLUSIONS Based on our results, the fecal microbiota composition at the age of 9 and 12 months is not associated with the development of CD. Our results, however, do not exclude the possibility of duodenal microbiota changes or a later microbiota-related trigger for the disease.
Collapse
Affiliation(s)
- Anniina Rintala
- a Department of Medical Microbiology and Immunology , University of Turku , Turku , Finland.,b Department of Clinical Microbiology , Turku University Hospital , Turku , Finland
| | - Iiris Riikonen
- a Department of Medical Microbiology and Immunology , University of Turku , Turku , Finland
| | - Anne Toivonen
- c Department of Bacteriology and Immunology , University of Helsinki and Laboratory Services (HUSLAB), Division of Clinical Microbiology, Helsinki University Hospital , Helsinki , Finland.,d Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Sami Pietilä
- e Turku Centre for Biotechnology , University of Turku and Åbo Akademi University , Turku , Finland
| | - Eveliina Munukka
- a Department of Medical Microbiology and Immunology , University of Turku , Turku , Finland
| | | | - Laura L Elo
- e Turku Centre for Biotechnology , University of Turku and Åbo Akademi University , Turku , Finland
| | - Pekka Arikoski
- g Department of Pediatrics , Kuopio University Hospital and University of Eastern Finland , Kuopio , Finland
| | - Kristiina Luopajärvi
- h Children's Hospital, Department of Pediatrics , Helsinki University Hospital and University of Helsinki , Helsinki , Finland
| | - Ursula Schwab
- i Institute of Public Health and Clinical Nutrition , University of Eastern Finland , Kuopio , Finland
| | - Matti Uusitupa
- i Institute of Public Health and Clinical Nutrition , University of Eastern Finland , Kuopio , Finland
| | - Seppo Heinonen
- j Department of Obstetrics and Gynecology , Helsinki University Hospital and University of Helsinki , Helsinki , Finland
| | - Erkki Savilahti
- h Children's Hospital, Department of Pediatrics , Helsinki University Hospital and University of Helsinki , Helsinki , Finland
| | - Erkki Eerola
- a Department of Medical Microbiology and Immunology , University of Turku , Turku , Finland.,b Department of Clinical Microbiology , Turku University Hospital , Turku , Finland
| | - Jorma Ilonen
- a Department of Medical Microbiology and Immunology , University of Turku , Turku , Finland.,k Immunogenetics Laboratory , Institute of Biomedicine, University of Turku , Turku , Finland
| |
Collapse
|
50
|
Olivares M, Walker AW, Capilla A, Benítez-Páez A, Palau F, Parkhill J, Castillejo G, Sanz Y. Gut microbiota trajectory in early life may predict development of celiac disease. MICROBIOME 2018; 6:36. [PMID: 29458413 PMCID: PMC5819212 DOI: 10.1186/s40168-018-0415-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/24/2018] [Indexed: 05/28/2023]
Abstract
BACKGROUND To investigate whether alterations in the developing intestinal microbiota and immune markers precede celiac disease (CD) onset in infants at familial risk of developing the disease. METHODS A nested case-control study was carried out as part of a larger prospective cohort study, which included healthy full-term newborns (> 200) with at least one first relative with biopsy-verified CD. The present study includes cases of CD (n = 10) and the best-matched controls (n = 10) who did not develop the disease after 5-year follow-up. Fecal microbiota, assessed by high-throughput 16S rRNA gene amplicon sequencing, and immune parameters were profiled at 4 and 6 months of age and related to CD onset. RESULTS The microbiota of infants who remained healthy showed an increase in bacterial diversity over time, characterized by increases in Firmicutes families, but not those who developed CD. Infants who subsequently developed CD showed a significant reduction in sIgA levels over time, while those who remained healthy showed increases in TNF-α correlated to Bifidobacterium spp. An increased relative abundance of Bifidobacterium longum was associated with control children while increased proportions of Bifidobacterium breve and Enterococcus spp. were associated with CD development. CONCLUSION The findings suggest that alterations in the early trajectory of gut microbiota in infants at CD risk could influence the immune maturation process and predispose to CD, although larger population studies are warranted to confirm this hypothesis.
Collapse
Affiliation(s)
- Marta Olivares
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/Catedrático Agustín Escardino, 7. 46980, Paterna, Valencia, Spain
| | - Alan W. Walker
- Gut Health Group, The Rowett Institute, University of Aberdeen, Aberdeen, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire UK
| | - Amalia Capilla
- Genetics and Molecular Medicine Unit, Institute of Biomedicine of Valencia, National Research Council (IBV-CSIC), Valencia, Spain
- Center for regenerative medicine, Boston university school of medicine, Boston, USA
| | - Alfonso Benítez-Páez
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/Catedrático Agustín Escardino, 7. 46980, Paterna, Valencia, Spain
| | - Francesc Palau
- Genetics and Molecular Medicine Unit, Institute of Biomedicine of Valencia, National Research Council (IBV-CSIC), Valencia, Spain
- Institut de Recerca Sant Joan de Déu and CIBERER, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Gemma Castillejo
- Hospital Universitari de Sant Joan de Reus, IISPV, URV, Tarragona, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/Catedrático Agustín Escardino, 7. 46980, Paterna, Valencia, Spain
| |
Collapse
|