1
|
de Oliveira Matos A, Henrique Dos Santos Dantas P, Rodrigues do Carmo Neto J, Contreras Colmenares MT, Felice AG, de Castro Soares S, Silva-Sales M, Sales-Campos H. Uncovering the role of TREM-1 in celiac disease: In silico insights into the recognition of gluten-derived peptides and inflammatory mechanisms. Comput Biol Med 2025; 189:109981. [PMID: 40056844 DOI: 10.1016/j.compbiomed.2025.109981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/31/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Celiac disease (CD) is a chronic enteropathy characterized by a permanent intolerance to gluten. While CD has been associated with heightened T cell responses and the involvement of distinct innate immunity components, the role of the triggering receptor expressed on myeloid cells (TREM) family in this disease remains unclear. Thus, as TREM-1 has already been implicated in other inflammatory bowel diseases, and given its role in the amplification of inflammation, we hypothesized that it might play a role in the pathophysiology of CD. METHODS AND RESULTS the STRING tool was used to predict protein-protein interaction networks between TREM-1 and CD signaling pathways. Then, molecular docking and molecular dynamics simulations were conducted to explore potential interactions between TREM-1 and different peptides derived from alpha-gliadin (25-mer, 33-mer and p31-43). Finally, we used transcriptomic data, available from public repositories, to assess TREM1 gene expression, and genes involved in its signaling pathway, in CD patients. Our results found an association between TREM-1 and CD markers, with STRING analysis, and the in silico simulations suggesting that the receptor might recognize the alpha-gliadin peptides, with the TREM-1/p31-43 interaction as the most likely interaction to occur biologically. Furthermore, TREM1 and its signaling pathway were increased in patients with active CD, while in those in clinical remission, the expression levels were similar to healthy controls. CONCLUSIONS collectively, our findings suggest that TREM-1 might recognize alpha-gliadin derived peptides, and TREM-1's activation may contribute to the intestinal inflammation observed in CD.
Collapse
Affiliation(s)
- Amanda de Oliveira Matos
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil.
| | | | | | | | - Andrei Giacchetto Felice
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Siomar de Castro Soares
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Marcelle Silva-Sales
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil.
| | | |
Collapse
|
2
|
Bascuñán KA, Araya M, Rodríguez JM, Roncoroni L, Elli L, Alvarez JDPL, Valenzuela R. Interplay of n-3 Polyunsaturated Fatty Acids, Intestinal Inflammation, and Gut Microbiota in Celiac Disease Pathogenesis. Nutrients 2025; 17:621. [PMID: 40004950 PMCID: PMC11858531 DOI: 10.3390/nu17040621] [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: 01/03/2025] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Celiac disease (CD) is a chronic autoimmune disorder driven by both genetic and environmental factors, with the HLA DQ2/DQ8 genotypes playing a central role in its development. Despite the genetic predisposition, only a small percentage of individuals carrying these genotypes develop the disease. Gluten, a protein found in wheat, rye, and barley, is the primary environmental trigger, but other factors, such as the intestinal microbiota, may also contribute to disease progression. While the gluten-free diet (GFD) remains the cornerstone of treatment, many CD patients experience persistent inflammation and gut dysbiosis, leading to ongoing symptoms and complications. This chronic inflammation, which impairs nutrient absorption, increases the risk of malnutrition, anemia, and other autoimmune disorders. Recent studies have identified an altered gut microbiota in CD patients, both on and off the GFD, highlighting the potential role of the microbiota in disease pathogenesis. An emerging area of interest is the supplementation of n-3 polyunsaturated fatty acids (PUFAs), known for their anti-inflammatory properties, as a potential therapeutic strategy. n-3 PUFAs, found in fish oil and certain plant oils, modulate the immune cell function and cytokine production, making them a promising intervention for controlling chronic inflammation in CD. This review explores the current understanding of n-3 PUFAs' effects on the gut microbiota's composition and inflammation in CD, with the goal of identifying new avenues for complementary treatments to improve disease management.
Collapse
Affiliation(s)
- Karla A. Bascuñán
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| | - Magdalena Araya
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Juan Manuel Rodríguez
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | | | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| |
Collapse
|
3
|
Ogulur I, Pat Y, Yazici D, Ardicli S, Ardicli O, Mitamura Y, Akdis M, Akdis CA. Epithelial barrier dysfunction, type 2 immune response, and the development of chronic inflammatory diseases. Curr Opin Immunol 2024; 91:102493. [PMID: 39321494 DOI: 10.1016/j.coi.2024.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, humans have been increasingly exposed to substances toxic for epithelial cells, including air pollutants, laundry and dishwashers, household chemicals, toothpaste, food additives, microplastics, and nanoparticles, introduced into our daily lives as part of industrialization, urbanization, and modernization. These substances disrupt the epithelial barriers and lead to microbial dysbiosis and cause immune response to allergens, opportunistic pathogens, bacterial toxins, and autoantigens followed by chronic inflammation due to epigenetic mechanisms. Recent evidence from studies on the mechanisms of epithelial barrier damage has demonstrated that even trace amounts of toxic substances can damage epithelial barriers and induce tissue inflammation. Further research in this field is essential for our understanding of the causal substances and molecular mechanisms involved in the initiation of leaky epithelial barriers that cascade into chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
4
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Kistler W, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024; 79:3192-3237. [PMID: 39370939 DOI: 10.1111/all.16318] [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: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Walter Kistler
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
5
|
Luz VCC, Pereira SG. Celiac disease gut microbiome studies in the third millennium: reviewing the findings and gaps of available literature. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 6:1413637. [PMID: 39355139 PMCID: PMC11444026 DOI: 10.3389/fmedt.2024.1413637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/29/2024] [Indexed: 10/03/2024] Open
Abstract
Celiac disease is an autoimmune enteropathy caused by the ingestion of minute amounts of gluten in a subset of genetically predisposed individuals. Its onset occurs at different ages and with variable symptoms. The gut microbiome may contribute to this variability. This review aims to provide an overview of the available research on celiac disease gut microbiome and identify the knowledge gap that could guide future studies. Following the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-analysis extension for Scoping Reviews (PRISMA-ScR), four electronic databases were searched for literature from January 2000 to July 2023 addressing celiac disease gut microbiome characterization using next-generation sequencing (NGS) approaches. From the 489 publications retrieved, 48 publications were selected and analyzed, focusing on sample characterization (patients, controls, and tissues) and methodologies used for NGS microbiome analysis and characterization. The majority of the selected publications regarded children and adults, and four were randomized clinical trials. The number of participants per study greatly varied and was typically low. Feces were the most frequently tested sample matrix, and duodenal samples were analyzed in one-third of the studies. Incomplete and diverse information on the methodological approaches and gut microbiome results was broadly observed. While similar trends regarding the relative abundance of some phyla, such as Pseudomonadota (former Proteobacteria), were detected in some studies, others contradicted those results. The observed high variability of technical approaches and possibly low power and sample sizes may prevent reaching a consensus on celiac disease gut microbiome composition. Standardization of research protocols to allow reproducibility and comparability is required, as interdisciplinary collaborations to further data analysis, interpretation, and, more importantly, health outcome prediction or improvement.
Collapse
Affiliation(s)
| | - Sónia Gonçalves Pereira
- Center for Innovative Care and Health Technology, School of Health Sciences, Polytechnic of Leiria, Leiria, Portugal
| |
Collapse
|
6
|
Hummel G, Aagaard K. Arthropods to Eutherians: A Historical and Contemporary Comparison of Sparse Prenatal Microbial Communities Among Animalia Species. Am J Reprod Immunol 2024; 92:e13897. [PMID: 39140417 DOI: 10.1111/aji.13897] [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: 04/24/2023] [Revised: 04/08/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Since the advent of next-generation sequencing, investigators worldwide have sought to discern whether a functional and biologically or clinically relevant prenatal microbiome exists. One line of research has led to the hypothesis that microbial DNA detected in utero/in ovo or prior to birth/hatching is a result of contamination and does not belong to viable and functional microbes. Many of these preliminary evaluations have been conducted in humans, mice, and nonhuman primates due to sample and specimen availability. However, a comprehensive review of the literature across animal species suggests organisms that maintain an obligate relationship with microbes may act as better models for interrogating the selective pressures placed on vertical microbial transfer over traditional laboratory species. To date, studies in humans and viviparous laboratory species have failed to illustrate the clear presence and transfer of functional microbes in utero. Until a ground truth regarding the status and relevance of prenatal microbes can be ascertained, it is salient to conduct parallel investigations into the prevalence of a functional prenatal microbiome across the developmental lifespan of multiple organisms in the kingdom Animalia. This comprehensive understanding is necessary not only to determine the role of vertically transmitted microbes and their products in early human health but also to understand their full One Health impact. This review is among the first to compile such comprehensive primary conclusions from the original investigator's conclusions, and hence collectively illustrates that prenatal microbial transfer is supported by experimental evidence arising from over a long and rigorous scientific history encompassing a breadth of species from kingdom Animalia.
Collapse
Affiliation(s)
- Gwendolynn Hummel
- Departments of Obstetrics and Gynecology (Division of Maternal-Fetal Medicine) and Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Kjersti Aagaard
- Departments of Obstetrics and Gynecology (Division of Maternal-Fetal Medicine) and Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
7
|
Rostami-Nejad M, Asri N, Bakhtiari S, Khalkhal E, Maleki S, Rezaei-Tavirani M, Jahani-Sherafat S, Rostami K. Metabolomics and lipidomics signature in celiac disease: a narrative review. Clin Exp Med 2024; 24:34. [PMID: 38340186 PMCID: PMC10858823 DOI: 10.1007/s10238-024-01295-2] [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: 06/06/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Celiac disease (CD) is a chronic immune-mediated inflammatory disease of the small intestine caused by aberrant immune responses to consumed gluten proteins. CD is diagnosed by a combination of the patients reported symptoms, serologic and endoscopic biopsy evaluation of the small intestine; and adherence to a strict gluten-free diet (GFD) is considered the only available therapeutic approach for this disorder. Novel approaches need to be considered for finding new biomarkers to help this disorder diagnosis and finding a new alternative therapeutic method for this group of patients. Metabolomics and lipidomics are powerful tools to provide highly accurate and sensitive biomarkers. Previous studies indicated a metabolic fingerprint for CD deriving from alterations in gut microflora or intestinal permeability, malabsorption, and energy metabolism. Moreover, since CD is characterized by increased intestinal permeability and due to the importance of membrane lipid components in controlling barrier integrity, conducting lipidomics studies in this disorder is of great importance. In the current study, we tried to provide a critical overview of metabolomic and lipidomic changes in CD.
Collapse
Affiliation(s)
- Mohammad Rostami-Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nastaran Asri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Bakhtiari
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Khalkhal
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Maleki
- Department of Computer Science, University of Tabriz, Tabriz, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Jahani-Sherafat
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rostami
- Department of Gastroenterology, MidCentral DHB, Palmerston North, 4442, New Zealand
| |
Collapse
|
8
|
Lauko S, Gancarcikova S, Hrckova G, Hajduckova V, Andrejcakova Z, Fecskeova LK, Bertkova I, Hijova E, Kamlarova A, Janicko M, Ambro L, Kvakova M, Gulasova Z, Strojny L, Strkolcova G, Mudronova D, Madar M, Demeckova V, Nemetova D, Pacuta I, Sopkova D. Beneficial Effect of Faecal Microbiota Transplantation on Mild, Moderate and Severe Dextran Sodium Sulphate-Induced Ulcerative Colitis in a Pseudo Germ-Free Animal Model. Biomedicines 2023; 12:43. [PMID: 38255150 PMCID: PMC10813722 DOI: 10.3390/biomedicines12010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Transplantation of faecal microbiota (FMT) is generally considered a safe therapeutic procedure with few adverse effects. The main factors that limit the spread of the use of FMT therapy for idiopathic inflammatory bowel disease (IBD) are the necessity of minimising the risk of infection and transfer of another disease. Obtaining the animal model of UC (ulcerative colitis) by exposure to DSS (dextran sodium sulphate) depends on many factors that significantly affect the result. Per os intake of DSS with water is individual for each animal and results in the development of a range of various forms of induced UC. For this reason, the aim of our study was to evaluate the modulation and regenerative effects of FMT on the clinical and histopathological responses and the changes in the bowel microenvironment in pseudo germ-free (PGF) mice of the BALB/c line subjected to chemical induction of mild, moderate and serious forms of UC. The goal was to obtain new data related to the safety and effectiveness of FMT that can contribute to its improved and optimised use. The animals with mild and moderate forms of UC subjected to FMT treatment exhibited lower severity of the disease and markedly lower damage to the colon, including reduced clinical and histological disease index and decreased inflammatory response of colon mucosa. However, FMT treatment failed to achieve the expected therapeutic effect in animals with the serious form of UC activity. The results of our study indicated a potential safety risk involving development of bacteraemia and also translocation of non-pathogenic representatives of bowel microbiota associated with FMT treatment of animals with a diagnosed serious form of UC.
Collapse
Affiliation(s)
- Stanislav Lauko
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Sona Gancarcikova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Gabriela Hrckova
- Institute of Parasitology, Slovak Academy of Sciences, 041 81 Kosice, Slovakia;
| | - Vanda Hajduckova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Zuzana Andrejcakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (Z.A.); (D.S.)
| | - Livia Kolesar Fecskeova
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital (UHLP) in Kosice, 040 11 Kosice, Slovakia;
| | - Izabela Bertkova
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Emilia Hijova
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Anna Kamlarova
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Martin Janicko
- 2nd Department of Internal Medicine, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital in Kosice, 040 11 Kosice, Slovakia;
| | - Lubos Ambro
- Center for Interdisciplinary Biosciences, Technology and Innovation Park, Pavol Jozef Safarik University in Kosice, 040 01 Kosice, Slovakia;
| | - Monika Kvakova
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Zuzana Gulasova
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Ladislav Strojny
- Center of Clinical and Preclinical Research—MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia; (I.B.); (E.H.); (A.K.); (M.K.); (Z.G.); (L.S.)
| | - Gabriela Strkolcova
- Department of Epizootiology, Parasitology and Protection of One Health, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia;
| | - Dagmar Mudronova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Marian Madar
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Vlasta Demeckova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, 040 01 Kosice, Slovakia;
| | - Daniela Nemetova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Ivan Pacuta
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (S.L.); (V.H.); (D.M.); (M.M.); (D.N.); (I.P.)
| | - Drahomira Sopkova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia; (Z.A.); (D.S.)
| |
Collapse
|
9
|
Zhang D, Zheng Y, Wang X, Wang D, Luo H, Zhu W, Zhang W, Chen Z, Shao J. Effects of Dietary Fish Meal Replaced by Fish Steak Meal on Growth Performance, Antioxidant Capacity, Intestinal Health and Microflora, Inflammatory Response, and Protein Metabolism of Large Yellow Croaker Larimichthys crocea. AQUACULTURE NUTRITION 2023; 2023:2733234. [PMID: 38152156 PMCID: PMC10752682 DOI: 10.1155/2023/2733234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/04/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023]
Abstract
Although fish steak meal (FSM) is a potentially available protein source, its efficiency as a fish meal (FM) substitute remains unclear to date. To this end, this study was carried out to determine the effects of dietary FM replaced by FSM on growth performance, antioxidant capacity, intestinal health and microflora, inflammatory response, and protein metabolism of large yellow croaker. Five isolipidic and isonitrogenous diets were formulated by substituting FM with FSM at levels of 0% (FSM0, control diet), 25% (FSM25), 50% (FSM50), 75% (FSM75), and 100% (FSM100), and were fed to juvenile large yellow croaker for 8 weeks. Compared with the control diet, the replacement of 25% dietary FM with FSM did not markedly alter the weight gain (WG) and specific growth rate (SGR). When the FM substitution level was over 25%, WG and SGR markedly reduced. The intestinal structure observation found that the FSM75 and FSM100 diets markedly decreased villus height, villus width, and muscle thickness of the anterior intestine. The FSM75 and FSM100 diets significantly decreased enzyme activities of amylase (AMS), lipase (LPS), trypsin, catalase (CAT), and total superoxide dismutase (T-SOD) and the total antioxidant capacity (T-AOC), and increased the malondialdehyde (MDA) content in the liver of large yellow croaker. The mRNA expression levels of intestinal barrier and inflammatory response-related genes suggested that the FSM50, FSM75, and FSM100 diets significantly decreased the mRNA abundances of intestinal barrier-related genes and anti-inflammatory response-related genes, and increased the mRNA abundances of proinflammatory gene il-6 in the anterior intestine. The compositions of intestinal microflora displayed that the FSM50, FSM75, and FSM100 diets decreased relative abundances of Firmicutes phylum and increased relative abundances of Proteobacteria phylum. In addition, the results of protein expression levels showed that the phosphorylation level of mammalian target of rapamycin (mTOR) and 4E-binding protein 1 (4E-BP1) in FSM75 and FSM100 groups were markedly reduced. In conclusion, FSM can replace up to 25% dietary FM without compromising the growth performance, intestinal health, and protein metabolism of the large yellow croaker.
Collapse
Affiliation(s)
- Dianguang Zhang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yunzong Zheng
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xuexi Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Dejuan Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hongjie Luo
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenbo Zhu
- Fuzhou Haima Feed Co. Ltd., Fuzhou 350311, China
| | - Weini Zhang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | | | - Jianchun Shao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fuzhou Institute of Oceanography, Fuzhou 350108, China
| |
Collapse
|
10
|
Tosi M, Montanari C, Bona F, Tricella C, Agostinelli M, Dolor J, Chillemi C, Di Profio E, Tagi VM, Vizzuso S, Fiore G, Zuccotti G, Verduci E. Dietary Inflammatory Potential in Pediatric Diseases: A Narrative Review. Nutrients 2023; 15:5095. [PMID: 38140353 PMCID: PMC10745369 DOI: 10.3390/nu15245095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammatory status is one of the main drivers in the development of non-communicable diseases (NCDs). Specific unhealthy dietary patterns and the growing consumption of ultra-processed foods (UPFs) may influence the inflammation process, which negatively modulates the gut microbiota and increases the risk of NCDs. Moreover, several chronic health conditions require special long-term dietary treatment, characterized by altered ratios of the intake of nutrients or by the consumption of disease-specific foods. In this narrative review, we aimed to collect the latest evidence on the pro-inflammatory potential of dietary patterns, foods, and nutrients in children affected by multifactorial diseases but also on the dietetic approaches used as treatment for specific diseases. Considering multifactorial diet-related diseases, the triggering effect of pro-inflammatory diets has been addressed for metabolic syndrome and inflammatory bowel diseases, and the latter for adults only. Future research is required on multiple sclerosis, type 1 diabetes, and pediatric cancer, in which the role of inflammation is emerging. For diseases requiring special diets, the role of single or multiple foods, possibly associated with inflammation, was assessed, but more studies are needed. The evidence collected highlighted the need for health professionals to consider the entire dietary pattern, providing balanced and healthy diets not only to permit the metabolic control of the disease itself, but also to prevent the development of NCDs in adolescence and adulthood. Personalized nutritional approaches, in close collaboration between the hospital, country, and families, must always be promoted together with the development of new methods for the assessment of pro-inflammatory dietary habits in pediatric age and the implementation of telemedicine.
Collapse
Affiliation(s)
- Martina Tosi
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Health Sciences, University of Milan, 20146 Milan, Italy;
| | - Chiara Montanari
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milan, 20157 Milan, Italy
| | - Federica Bona
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Chiara Tricella
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Marta Agostinelli
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Jonabell Dolor
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Claudia Chillemi
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Health Sciences, University of Milan, 20146 Milan, Italy;
| | - Veronica Maria Tagi
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Health Sciences, University of Milan, 20146 Milan, Italy;
| | - Sara Vizzuso
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Health Sciences, University of Milan, 20146 Milan, Italy;
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.T.); (C.M.); (F.B.); (C.T.); (M.A.); (J.D.); (C.C.); (E.D.P.); (V.M.T.); (S.V.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milan, 20157 Milan, Italy
| | - Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy;
- Metabolic Diseases Unit, Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy
| |
Collapse
|
11
|
Mozafarybazargany M, Khonsari M, Sokoty L, Ejtahed HS, Qorbani M. The effects of probiotics on gastrointestinal symptoms and microbiota in patients with celiac disease: a systematic review and meta-analysis on clinical trials. Clin Exp Med 2023; 23:2773-2788. [PMID: 36609792 DOI: 10.1007/s10238-022-00987-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/30/2022] [Indexed: 01/08/2023]
Abstract
Gluten-free diet (GFD) is the most effective method to manage celiac disease (CD). Many patients do not reach the complete symptom alleviation, even by strict GFD. Recent studies have reported inconsistent results regarding the beneficial benefits of taking probiotics. Therefore, we aimed to evaluate the effects of probiotics on gastrointestinal (GI) symptoms and the possible underlying causes in CD and celiac disease autoimmunity (CDA) patients. Databases, including PubMed, Scopus, Embase, Web of Science and Google Scholar, were searched for clinical trials published until July 2022 about assessing the effects of probiotics or synbiotics on CD or CDA patients. We collected data on GI symptoms, CD markers, inflammatory and immune responses, adverse events, and gut microbiota. A random effect meta-analysis was used to estimate the pooled standardized mean difference (SMD) and confidence interval (CI). We screened 7234 articles, of which 14 were included in the qualitative analysis and 5 in the quantitative analysis. Probiotics might alleviate GI symptoms, especially in the highly symptomatic patients, and improve immune response in CD and CDA patients. Results of the meta-analysis showed that probiotics increased the abundance of Bifidobacterium (SMD: 0.72, 95%CI (0.13, 1.30) and Lactobacillus (SMD: 0.49, 95%CI (0.18, 0.80) as compared with placebo. Probiotics did not increase the adverse events compared to the placebo. Probiotics might alleviate GI symptoms and immune response and improve dysbiosis in CD and CDA patients. However, high-quality clinical trials are needed to increase the level of evidence. Also, the most suitable combination of probiotics is yet to find.
Collapse
Affiliation(s)
| | - Mohammadian Khonsari
- Social Determinants of Health Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Leily Sokoty
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mostafa Qorbani
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Skoracka K, Hryhorowicz S, Rychter AM, Ratajczak AE, Szymczak-Tomczak A, Zawada A, Słomski R, Dobrowolska A, Krela-Kaźmierczak I. Why are western diet and western lifestyle pro-inflammatory risk factors of celiac disease? Front Nutr 2023; 9:1054089. [PMID: 36742009 PMCID: PMC9895111 DOI: 10.3389/fnut.2022.1054089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/31/2022] [Indexed: 01/21/2023] Open
Abstract
The prevalence of celiac disease increased in recent years. In addition to the genetic and immunological factors, it appears that environmental determinants are also involved in the pathophysiology of celiac disease. Gastrointestinal infections impact the development of celiac disease. Current research does not directly confirm the protective effect of natural childbirth and breastfeeding on celiac disease. However, it seems that in genetically predisposed children, the amount of gluten introduced into the diet may have an impact on celiac disease development. Also western lifestyle, including western dietary patterns high in fat, sugar, and gliadin, potentially may increase the risk of celiac disease due to changes in intestinal microbiota, intestinal permeability, or mucosal inflammation. Further research is needed to expand the knowledge of the relationship between environmental factors and the development of celiac disease to define evidence-based preventive interventions against the development of celiac disease. The manuscript summarizes current knowledge on factors predisposing to the development of celiac disease including factors associated with the western lifestyle.
Collapse
Affiliation(s)
- Kinga Skoracka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland,Doctoral School, Poznan University of Medical Sciences, Poznań, Poland,*Correspondence: Kinga Skoracka ✉
| | | | - Anna Maria Rychter
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland,Doctoral School, Poznan University of Medical Sciences, Poznań, Poland
| | - Alicja Ewa Ratajczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland,Doctoral School, Poznan University of Medical Sciences, Poznań, Poland
| | - Aleksandra Szymczak-Tomczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Agnieszka Zawada
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Iwona Krela-Kaźmierczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
13
|
Shi T, Feng Y, Liu W, Liu H, Li T, Wang M, Li Z, Lu J, Abudurexiti A, Maimaitireyimu A, Hu J, Gao F. Characteristics of gut microbiota and fecal metabolomes in patients with celiac disease in Northwest China. Front Microbiol 2022; 13:1020977. [PMID: 36519162 PMCID: PMC9742481 DOI: 10.3389/fmicb.2022.1020977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 06/30/2024] Open
Abstract
Celiac disease (CD) is an autoimmune small bowel disease. The pattern of gut microbiota is closely related to dietary habits, genetic background, and geographical factors. There is a lack of research on CD-related gut microbiota in China. This study aimed to use 16S rDNA sequencing and metabolomics to analyze the fecal microbial composition and metabolome characteristics in patients diagnosed with CD in Northwest China, and to screen potential biomarkers that could be used for its diagnosis. A significant difference in the gut microbiota composition was observed between the CD and healthy controls groups. At the genus level, the abundance of Streptococcus, Lactobacillus, Veillonella, and Allisonella communities in the CD group were increased (Q < 0.05). Furthermore, the abundance of Ruminococcus, Faecalibacterium, Blautia, Gemmiger, and Anaerostipes community in this group were decreased (Q < 0.05). A total of 222 different fecal metabolites were identified in the two groups, suggesting that CD patients have a one-carbon metabolism defect. Four species of bacteria and six metabolites were selected as potential biomarkers using a random forest model. Correlation analysis showed that changes in the gut microbiota were significantly correlated with changes in fecal metabolite levels. In conclusion, the patterns of distribution of gut microbiota and metabolomics in patients with CD in Northwest China were found to be unique to these individuals. This has opened up a new way to explore potential beneficial effects of supplementing specific nutrients and potential diagnostic and therapeutic targets in the future.
Collapse
Affiliation(s)
- Tian Shi
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Yan Feng
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Weidong Liu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Huan Liu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ting Li
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Man Wang
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ziqiong Li
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Jiajie Lu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Adilai Abudurexiti
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ayinuer Maimaitireyimu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Jiali Hu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Feng Gao
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| |
Collapse
|
14
|
Mahalak KK, Firrman J, Bobokalonov J, Narrowe AB, Bittinger K, Daniel S, Tanes C, Mattei LM, Zeng WB, Soares JW, Kobori M, Lemons JMS, Tomasula PM, Liu L. Persistence of the Probiotic Lacticaseibacillus rhamnosus Strain GG (LGG) in an In Vitro Model of the Gut Microbiome. Int J Mol Sci 2022; 23:12973. [PMID: 36361763 PMCID: PMC9657340 DOI: 10.3390/ijms232112973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/26/2023] Open
Abstract
The consumption of probiotics is widely encouraged due to reports of their positive effects on human health. In particular, Lacticaseibacillus rhamnosus strain GG (LGG) is an approved probiotic that has been reported to improve health outcomes, especially for gastrointestinal disorders. However, how LGG cooperates with the gut microbiome has not been fully explored. To understand the interaction between LGG and its ability to survive and grow within the gut microbiome, this study introduced LGG into established microbial communities using an in vitro model of the colon. LGG was inoculated into the simulated ascending colon and its persistence in, and transit through the subsequent transverse and descending colon regions was monitored over two weeks. The impact of LGG on the existing bacterial communities was investigated using 16S rRNA sequencing and short-chain fatty acid analysis. LGG was able to engraft and proliferate in the ascending region for at least 10 days but was diminished in the transverse and descending colon regions with little effect on short-chain fatty acid abundance. These data suggest that the health benefits of the probiotic LGG rely on its ability to transiently engraft and modulate the host microbial community.
Collapse
Affiliation(s)
- Karley K. Mahalak
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Jenni Firrman
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Jamshed Bobokalonov
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Adrienne B. Narrowe
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Scott Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lisa M. Mattei
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Wei-Bin Zeng
- Department of Mathematics, University of Louisville, Louisville, KY 40292, USA
| | - Jason W. Soares
- Bioprocessing and Bioengineering Group, US Army Combat Capabilities Development Command Soldier Center (CCDC-SC), Natick, MA 01760, USA
| | - Masuko Kobori
- Institute of Food Research, National Agriculture and Food Research Organization, Tsukuba 305-8642, Ibaraki, Japan
| | - Johanna M. S. Lemons
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Peggy M. Tomasula
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| | - LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, 600 E Mermaid Lane, Wyndmoor, PA 19038, USA
| |
Collapse
|
15
|
Wagh SK, Lammers KM, Padul MV, Rodriguez-Herrera A, Dodero VI. Celiac Disease and Possible Dietary Interventions: From Enzymes and Probiotics to Postbiotics and Viruses. Int J Mol Sci 2022; 23:ijms231911748. [PMID: 36233048 PMCID: PMC9569549 DOI: 10.3390/ijms231911748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
Celiac Disease (CeD) is a chronic small intestinal immune-mediated enteropathy caused by the ingestion of dietary gluten proteins in genetically susceptible individuals. CeD is one of the most common autoimmune diseases, affecting around 1.4% of the population globally. To date, the only acceptable treatment for CeD is strict, lifelong adherence to a gluten-free diet (GFD). However, in some cases, GFD does not alter gluten-induced symptoms. In addition, strict adherence to a GFD reduces patients’ quality of life and is often a socio-economic burden. This narrative review offers an interdisciplinary overview of CeD pathomechanism and the limitations of GFD, focusing on current research on possible dietary interventions. It concentrates on the recent research on the degradation of gluten through enzymes, the modulation of the microbiome, and the different types of “biotics” strategies, from probiotics to the less explored “viromebiotics” as possible beneficial complementary interventions for CeD management. The final aim is to set the context for future research that may consider the role of gluten proteins and the microbiome in nutritional and non-pharmacological interventions for CeD beyond the sole use of the GFD.
Collapse
Affiliation(s)
- Sandip K. Wagh
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Department of Biochemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad 431004, India
| | | | - Manohar V. Padul
- Department of Biochemistry, The Institute of Science, Dr. Homi Bhabha State University, Mumbai 400032, India
| | | | - Veronica I. Dodero
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Correspondence:
| |
Collapse
|
16
|
Wagh SK, Lammers KM, Padul MV, Rodriguez-Herrera A, Dodero VI. Celiac Disease and Possible Dietary Interventions: From Enzymes and Probiotics to Postbiotics and Viruses. Int J Mol Sci 2022. [PMID: 36233048 DOI: 10.3390/ijms231911748.pmid:36233048;pmcid:pmc9569549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Celiac Disease (CeD) is a chronic small intestinal immune-mediated enteropathy caused by the ingestion of dietary gluten proteins in genetically susceptible individuals. CeD is one of the most common autoimmune diseases, affecting around 1.4% of the population globally. To date, the only acceptable treatment for CeD is strict, lifelong adherence to a gluten-free diet (GFD). However, in some cases, GFD does not alter gluten-induced symptoms. In addition, strict adherence to a GFD reduces patients' quality of life and is often a socio-economic burden. This narrative review offers an interdisciplinary overview of CeD pathomechanism and the limitations of GFD, focusing on current research on possible dietary interventions. It concentrates on the recent research on the degradation of gluten through enzymes, the modulation of the microbiome, and the different types of "biotics" strategies, from probiotics to the less explored "viromebiotics" as possible beneficial complementary interventions for CeD management. The final aim is to set the context for future research that may consider the role of gluten proteins and the microbiome in nutritional and non-pharmacological interventions for CeD beyond the sole use of the GFD.
Collapse
Affiliation(s)
- Sandip K Wagh
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Department of Biochemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad 431004, India
| | | | - Manohar V Padul
- Department of Biochemistry, The Institute of Science, Dr. Homi Bhabha State University, Mumbai 400032, India
| | | | - Veronica I Dodero
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| |
Collapse
|
17
|
Carreras J. Artificial Intelligence Analysis of Celiac Disease Using an Autoimmune Discovery Transcriptomic Panel Highlighted Pathogenic Genes including BTLA. Healthcare (Basel) 2022; 10:1550. [PMID: 36011206 PMCID: PMC9408070 DOI: 10.3390/healthcare10081550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
Celiac disease is a common immune-related inflammatory disease of the small intestine caused by gluten in genetically predisposed individuals. This research is a proof-of-concept exercise focused on using Artificial Intelligence (AI) and an autoimmune discovery gene panel to predict and model celiac disease. Conventional bioinformatics, gene set enrichment analysis (GSEA), and several machine learning and neural network techniques were used on a publicly available dataset (GSE164883). Machine learning and deep learning included C5, logistic regression, Bayesian network, discriminant analysis, KNN algorithm, LSVM, random trees, SVM, Tree-AS, XGBoost linear, XGBoost tree, CHAID, Quest, C&R tree, random forest, and neural network (multilayer perceptron). As a result, the gene panel predicted celiac disease with high accuracy (95-100%). Several pathogenic genes were identified, some of the immune checkpoint and immuno-oncology pathways. They included CASP3, CD86, CTLA4, FASLG, GZMB, IFNG, IL15RA, ITGAX, LAG3, MMP3, MUC1, MYD88, PRDM1, RGS1, etc. Among them, B and T lymphocyte associated (BTLA, CD272) was highlighted and validated at the protein level by immunohistochemistry in an independent series of cases. Celiac disease was characterized by high BTLA, expressed by inflammatory cells of the lamina propria. In conclusion, artificial intelligence predicted celiac disease using an autoimmune discovery gene panel.
Collapse
Affiliation(s)
- Joaquim Carreras
- Department of Pathology, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Japan
| |
Collapse
|
18
|
Vacca M, Porrelli A, Calabrese FM, Lippolis T, Iacobellis I, Celano G, Pinto D, Russo F, Giannelli G, De Angelis M. How Metabolomics Provides Novel Insights on Celiac Disease and Gluten-Free Diet: A Narrative Review. Front Microbiol 2022; 13:859467. [PMID: 35814671 PMCID: PMC9260055 DOI: 10.3389/fmicb.2022.859467] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/27/2022] [Indexed: 12/12/2022] Open
Abstract
Celiac disease (CD) is an inflammatory autoimmune disorder triggered by the ingestion of gluten from wheat and other cereals. Nowadays, its positive diagnosis is based on invasive approaches such as the histological examination of intestinal biopsies and positive serology screening of antibodies. After proven diagnosis, the only admissible treatment for CD individuals is strict life-long adherence to gluten-free diet (GFD), although it is not a conclusive therapy. Acting by different mechanisms and with different etiologies, both CD and GFD have a great impact on gut microbiota that result in a different taxa composition. Altered production of specific metabolites reflects these microbiota changes. In this light, the currently available literature reports some suggestions about the possible use of specific metabolites, detected by meta-omics analyses, as potential biomarkers for a CD non-invasive diagnosis. To highlight insights about metabolomics application in CD study, we conducted a narrative dissertation of selected original articles published in the last decade. By applying a systematic search, it clearly emerged how the metabolomic signature appears to be contradictory, as well as poorly investigated.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Porrelli
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Francesco Maria Calabrese,
| | - Tamara Lippolis
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Ilaria Iacobellis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project-HMPA, Giuliani SpA, Milan, Italy
| | - Francesco Russo
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
19
|
Varma S, Green PH, Krishnareddy S. Clinical Factors Associated With Positive Stool PCR for Gastrointestinal Pathogens in Celiac and Inflammatory Bowel Disease. J Clin Gastroenterol 2022; 56:e196-e202. [PMID: 34999647 DOI: 10.1097/mcg.0000000000001657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/23/2021] [Indexed: 12/10/2022]
Abstract
GOALS We aimed to identify the clinical factors of celiac disease (CeD) and inflammatory bowel disease (IBD) associated with positive stool gastrointestinal (GI) polymerase chain reaction (PCR) test. BACKGROUND Understanding the pattern of enteric infections in CeD and IBD may allow further insight into microbiome-mediated pathogeneses. STUDY This was a retrospective study of adult patients (age 18 y or above) with CeD and IBD at a large quaternary care institution. We identified patients with CeD or IBD who underwent stool GIPCR evaluation as outpatients (office visit or at endoscopy) between March 2015 and March 2019. Patients with a negative GIPCR test within the study time frame were randomly chosen as controls (1 : 1). The independent relationship between clinical characteristics and positive GIPCR was evaluated using multivariable logistic regression. RESULTS A total of 266 patients met criteria for the study, including 92 (35%) with CeD and 174 (65%) with IBD. On multivariable analysis of factors associated with positive GIPCR test, CeD patients were more likely to have diarrheal presentation of illness [odds ratio (OR): 2.61, 95% confidence interval (CI) 1.05-6.72], experience extraintestinal manifestations (OR: 2.49, 95% CI: 1.01-6.31), and practice a gluten-free diet for at least 5 years (OR: 4.00, 95% CI: 1.36-11.67), relative to those with a negative GIPCR test. IBD patients with positive GIPCR were more likely to be on corticosteroids (OR: 2.23, 95% CI: 1.02-5.4.84), experience extraintestinal manifestations (OR: 2.60, 95% CI: 1.22-5.53), and use proton-pump inhibitors (OR: 4.07, 95% CI: 1.69-9.77). CONCLUSIONS Intestinal infections in CeD and IBD are associated with important disease-specific characteristics.
Collapse
Affiliation(s)
- Sanskriti Varma
- Department of Medicine, New-York Presbyterian Columbia University Medical Center
| | - Peter H Green
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Suneeta Krishnareddy
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
20
|
Sample D, Fouhse J, King S, Huynh HQ, Dieleman LA, Willing BP, Turner J. Baseline Fecal Microbiota in Pediatric Patients With Celiac Disease Is Similar to Controls But Dissimilar After 1 Year on the Gluten-Free Diet. JPGN REPORTS 2021; 2:e127. [PMID: 37206457 PMCID: PMC10191547 DOI: 10.1097/pg9.0000000000000127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/02/2021] [Indexed: 05/21/2023]
Abstract
The objectives of this pilot study were to examine fecal microbiota composition of pediatric patients with celiac disease (CD) before and after a 1-year gluten-free diet (GFD) and to determine the association with symptoms and anti-tissue transglutaminase (aTTG) antibody. Methods Stool samples were obtained from pediatric patients with CD and from healthy controls. Patients were classified by the presence (diarrhea, abdominal pain, weight loss) or absence (asymptomatic, headache, fatigue, etc.) of typical CD gastrointestinal symptoms and by aTTG normalization post-GFD intervention (< 7 U/mL). Fecal microbial composition was measured using 16S ribosomal RNA gene amplicon sequencing of the V3-V4 region. Results At diagnosis, 13 of 22 patients with CD had typical gastrointestinal symptoms, the remaining patients having atypical or asymptomatic presentations. After a 1-year GFD, all symptomatic patients improved and 9 of 19 had normalized aTTG. Prior to GFD, no distinct microbial signature was observed between patients and controls (P = 0.39). Post-GFD, patients with CD had a unique microbial signature with reductions in known fiber-degrading bacteria, including Blautia, Dorea, Lactobacillus, and Prevotella compared with controls. Within the patients with CD, microbial composition was not associated with reported symptom presentation or aTTG normalization. Conclusions Pediatric patients with CD only had a unique microbial signature compared with healthy controls when placed on the GFD. These results suggest that pediatric patients with CD may not have a unique fecal microbial signature indicative of inherent dysbiosis, in contrast to that suggested for older patients. In children with CD, diet may play a role in shaping microbial composition more so than disease status.
Collapse
Affiliation(s)
- Dory Sample
- From the Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Janelle Fouhse
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Seema King
- Department of Medicine, Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Hien Q. Huynh
- From the Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Levinus A. Dieleman
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Benjamin P. Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Justine Turner
- From the Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
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: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.
Collapse
Affiliation(s)
- Anna Torun
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Anna Hupalowska
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beata Pyrzynska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Sitkin SI, Avalueva EB, Oreshko LS, Khavkin AI. Intestinal microbiota and dysbiosis in celiac disease. ROSSIYSKIY VESTNIK PERINATOLOGII I PEDIATRII (RUSSIAN BULLETIN OF PERINATOLOGY AND PEDIATRICS) 2021; 66:116-122. [DOI: 10.21508/1027-4065-2021-66-2-116-122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Affiliation(s)
- S. I. Sitkin
- Mechnikov North-Western State Medical University; Almazov National Medical Research Centre; State Research Institute of Highly Pure Biopreparations
| | | | | | - A. I. Khavkin
- Veltischev Research and Clinical Institute for Pediatrics at the Pirogov Russian National Research Medical University
| |
Collapse
|
23
|
Ferrari E, Monzani R, Saverio V, Gagliardi M, Pańczyszyn E, Raia V, Villella VR, Bona G, Pane M, Amoruso A, Corazzari M. Probiotics Supplements Reduce ER Stress and Gut Inflammation Associated with Gliadin Intake in a Mouse Model of Gluten Sensitivity. Nutrients 2021; 13:1221. [PMID: 33917155 PMCID: PMC8067866 DOI: 10.3390/nu13041221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Exposure to gluten, a protein present in wheat rye and barley, is the major inducer for human Celiac Disease (CD), a chronic autoimmune enteropathy. CD occurs in about 1% worldwide population, in genetically predisposed individuals bearing human leukocyte antigen (HLA) DQ2/DQ8. Gut epithelial cell stress and the innate immune activation are responsible for the breaking oral tolerance to gliadin, a gluten component. To date, the only treatment available for CD is a long-term gluten-free diet. Several studies have shown that an altered composition of the intestinal microbiota (dysbiosis) could play a key role in the pathogenesis of CD through the modulation of intestinal permeability and the regulation of the immune system. Here, we show that gliadin induces a chronic endoplasmic reticulum (ER) stress condition in the small intestine of a gluten-sensitive mouse model and that the coadministration of probiotics efficiently attenuates both the unfolded protein response (UPR) and gut inflammation. Moreover, the composition of probiotics formulations might differ in their activity at molecular level, especially toward the three axes of the UPR. Therefore, probiotics administration might potentially represent a new valuable strategy to treat gluten-sensitive patients, such as those affected by CD.
Collapse
Affiliation(s)
- Eleonora Ferrari
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Romina Monzani
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Valentina Saverio
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Mara Gagliardi
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Elżbieta Pańczyszyn
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, 80134 Naples, Italy;
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Gianni Bona
- Division of Pediatrics, Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Marco Pane
- Probiotical Research Srl, 28100 Novara, Italy; (M.P.); (A.A.)
| | - Angela Amoruso
- Probiotical Research Srl, 28100 Novara, Italy; (M.P.); (A.A.)
| | - Marco Corazzari
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
24
|
Warbeck C, Dowd AJ, Kronlund L, Parmar C, Daun JT, Wytsma-Fisher K, Millet GY, Schick A, Reimer RA, Fung T, Culos-Reed SN. Feasibility and effects on the gut microbiota of a 12-week high-intensity interval training plus lifestyle education intervention on inactive adults with celiac disease. Appl Physiol Nutr Metab 2021; 46:325-336. [DOI: 10.1139/apnm-2020-0459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study assessed the feasibility and benefits of high-intensity interval training (HIIT) plus lifestyle education among inactive adults with celiac disease. Forty-one participants were randomized to receive the intervention (HIIT plus lifestyle education; HIIT+) for 12 weeks or waitlist control (WLC). Testing was completed at baseline, immediately post-intervention, and 3 months post-intervention. Generalized estimating equations were used to assess changes in the outcome variables over time between the groups. Mean percent of age-predicted maximum heart rate was 97.9% and average rating of perceived exertion was 6.33 (out of 10) during HIIT intervals. Following the intervention, the HIIT+ showed enrichment in relative abundance of Parabacteroides and Defluviitaleaceae_UCG_011 while WLC showed enrichment in relative abundance of Roseburia intestinalis, Klebsiella, and Adlercreutzia. A unique set of taxa were differentially abundant between the groups at 3 months post-intervention. HIIT+ participants experienced a reduction in resting heart rate (−6.6 bpm) immediately post-intervention compared with WLC. Further research is needed to establish an optimal HIIT protocol that may improve maximal oxygen uptake and metabolic syndrome biomarkers. Findings from this pilot study provide preliminary evidence that an HIIT intervention is feasible for inactive adults with celiac disease and leads to favourable changes in resting heart rate alongside potentially beneficial shifts in gut microbiota. Trial registration number: ClinicalTrials.gov number NCT03520244. Novelty: HIIT leads to potentially beneficial changes in the gut microbiota of adults with celiac disease. An HIIT exercise intervention is feasible and well tolerated for patients with celiac disease.
Collapse
Affiliation(s)
- Cassandra Warbeck
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - A. Justine Dowd
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Liam Kronlund
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Candice Parmar
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Julia T. Daun
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Guillaume Y. Millet
- Univ Lyon, UJM-Saint-Etienne, Inter-university Laboratory of Human Movement Biology, EA 7424, F-42023, Saint-Etienne, France
| | - Alana Schick
- International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Raylene A. Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB T3B 6A8, Canada
| | - Tak Fung
- Research Computing Services, Information Technologies, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - S. Nicole Culos-Reed
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
25
|
Multidimensional Disadvantages of a Gluten-Free Diet in Celiac Disease: A Narrative Review. Nutrients 2021; 13:nu13020643. [PMID: 33669442 PMCID: PMC7920475 DOI: 10.3390/nu13020643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
A gluten-free diet is the mainstay method of treatment and the prevention of celiac disease complications. However, an inadequately balanced gluten-free diet can increase the risk of obesity, negatively affect glucose and lipid metabolism, and increase the risk of the metabolic syndrome. Therefore, an adequate nutritional counselling is necessary for patients diagnosed with celiac disease in order to prevent and treat the components of the metabolic syndrome.
Collapse
|
26
|
Singh P, Rawat A, Al-Jarrah B, Saraswathi S, Gad H, Elawad M, Hussain K, Hendaus MA, Al-Masri W, Malik RA, Al Khodor S, Akobeng AK. Distinctive Microbial Signatures and Gut-Brain Crosstalk in Pediatric Patients with Coeliac Disease and Type 1 Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22041511. [PMID: 33546364 PMCID: PMC7913584 DOI: 10.3390/ijms22041511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Coeliac disease (CD) and Type 1 diabetes mellitus (T1DM) are immune-mediated diseases. Emerging evidence suggests that dysbiosis in the gut microbiome plays a role in the pathogenesis of both diseases and may also be associated with the development of neuropathy. The primary goal in this cross-sectional pilot study was to identify whether there are distinct gut microbiota alterations in children with CD (n = 19), T1DM (n = 18) and both CD and T1DM (n = 9) compared to healthy controls (n = 12). Our second goal was to explore the relationship between neuropathy (corneal nerve fiber damage) and the gut microbiome composition. Microbiota composition was determined by 16S rRNA gene sequencing. Corneal confocal microscopy was used to determine nerve fiber damage. There was a significant difference in the overall microbial diversity between the four groups with healthy controls having a greater microbial diversity as compared to the patients. The abundance of pathogenic proteobacteria Shigella and E. coli were significantly higher in CD patients. Differential abundance analysis showed that several bacterial amplicon sequence variants (ASVs) distinguished CD from T1DM. The tissue transglutaminase antibody correlated significantly with a decrease in gut microbial diversity. Furthermore, the Bacteroidetes phylum, specifically the genus Parabacteroides was significantly correlated with corneal nerve fiber loss in the subjects with neuropathic damage belonging to the diseased groups. We conclude that disease-specific gut microbial features traceable down to the ASV level distinguish children with CD from T1DM and specific gut microbial signatures may be associated with small fiber neuropathy. Further research on the mechanisms linking altered microbial diversity with neuropathy are warranted.
Collapse
Affiliation(s)
- Parul Singh
- Research Department, Sidra Medicine, Doha 26999, Qatar or (P.S.); (A.R.); (B.A.-J.)
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 24404, Qatar
| | - Arun Rawat
- Research Department, Sidra Medicine, Doha 26999, Qatar or (P.S.); (A.R.); (B.A.-J.)
| | - Bara Al-Jarrah
- Research Department, Sidra Medicine, Doha 26999, Qatar or (P.S.); (A.R.); (B.A.-J.)
| | - Saras Saraswathi
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha 26999, Qatar; (S.S.); (M.E.); (W.A.-M.); (A.K.A.)
| | - Hoda Gad
- Department Medicine, Weill Cornell Medicine-Qatar, Doha 24144, Qatar; (H.G.); (R.A.M.)
| | - Mamoun Elawad
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha 26999, Qatar; (S.S.); (M.E.); (W.A.-M.); (A.K.A.)
| | - Khalid Hussain
- Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar;
| | | | - Wesam Al-Masri
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha 26999, Qatar; (S.S.); (M.E.); (W.A.-M.); (A.K.A.)
| | - Rayaz A. Malik
- Department Medicine, Weill Cornell Medicine-Qatar, Doha 24144, Qatar; (H.G.); (R.A.M.)
| | - Souhaila Al Khodor
- Research Department, Sidra Medicine, Doha 26999, Qatar or (P.S.); (A.R.); (B.A.-J.)
- Correspondence:
| | - Anthony K. Akobeng
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha 26999, Qatar; (S.S.); (M.E.); (W.A.-M.); (A.K.A.)
- Department Medicine, Weill Cornell Medicine-Qatar, Doha 24144, Qatar; (H.G.); (R.A.M.)
| |
Collapse
|
27
|
Abstract
Mucosal surfaces are distinctive sites exposed to environmental, dietary, and microbial antigens. Particularly in the gut, the host continuously actively adapts via complex interactions between the microbiota and dietary compounds and immune and other tissue cells. Regulatory T cells (Tregs) are critical for tuning the intestinal immune response to self- and non-self-antigens in the intestine. Its importance in intestinal homeostasis is illustrated by the onset of overt inflammation caused by deficiency in Treg generation, function, or stability in the gut. A substantial imbalance in Tregs has been observed in intestinal tissue during pathogenic conditions, when a tightly regulated and equilibrated system becomes dysregulated and leads to unimpeded and chronic immune responses. In this chapter, we compile and critically discuss the current knowledge on the key factors that promote Treg-mediated tolerance in the gut, such as those involved in intestinal Treg differentiation, specificity and suppressive function, and their immunophenotype during health and disease. We also discuss the current state of knowledge on Treg dysregulation in human intestine during pathological states such as inflammatory bowel disease (IBD), necrotizing enterocolitis (NEC), graft-versus-host disease (GVHD), and colorectal cancer (CRC), and how that knowledge is guiding development of Treg-targeted therapies to treat or prevent intestinal disorders.
Collapse
|
28
|
|
29
|
A link between appendectomy and gastrointestinal cancers: a large-scale population-based cohort study in Korea. Sci Rep 2020; 10:15670. [PMID: 32973258 PMCID: PMC7518248 DOI: 10.1038/s41598-020-72770-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
An association between appendectomy and subsequent gastrointestinal (GI) cancer development has been postulated, although the evidence is limited and inconsistent. To provide clarification, we investigated the link between appendectomy and GI cancers in a large nationwide appendectomy cohort. This cohort was derived from the claims database of the National Health Insurance Service in South Korea and comprised 158,101 patients who had undergone appendectomy between 2007 and 2014. A comparison cohort of 474,303 subjects without appendectomy was selected after 1:3 matching by age and sex. The incidence of GI cancers after appendectomy was observed, and risk factors for GI cancers were determined by using a multivariable-adjusted proportional hazards model. Appendectomy did not significantly increase the incidence of GI cancers in the overall population (1.529 and 1557 per 1000 person-years in the non-appendectomy and appendectomy cohorts, respectively). However, appendectomy significantly increased the incidence of GI cancers in subgroups consisting of elderly (≥ 60 years) patients (adjusted HR, 1.102; 95% confidence interval, 1.011-1.201; p = 0.028) or women (adjusted HR, 1.180; 95% confidence interval, 1.066-1.306; p = 0.001).
Collapse
|
30
|
Aaron L, Torsten M. Candida albicans in celiac disease: A wolf in sheep's clothing. Autoimmun Rev 2020; 19:102621. [PMID: 32693029 DOI: 10.1016/j.autrev.2020.102621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a commensal fungus with a potential pathogenicity and celiac disease is an autoimmune condition. Both share multiple pathophysiological junctions, including serological markers against cell-wall proteins of Candida, anti-gliadin antibodies are positive in both entities, gluten and a candidal virulence factor share sequence similarity and the autoantigen of celiac disease, the tissue transglutaminase, is pivotal in Candida albicans commensalism and hostile behavior and its covalently cross linked products are stable and resistant to breakdown in the two entities. Those autoimmune/infectious cross roads are the basis for the hypothesis that Candida albicans is an additional environmental factor for celiac disease autoimmunogenesis.
Collapse
|
31
|
Demiroren K. Can a Synbiotic Supplementation Contribute to Decreasing Anti-Tissue Transglutaminase Levels in Children with Potential Celiac Disease? Pediatr Gastroenterol Hepatol Nutr 2020; 23:397-404. [PMID: 32704500 PMCID: PMC7354869 DOI: 10.5223/pghn.2020.23.4.397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/22/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Synbiotics can alleviate some intestinal pathologies or prevent trigger mechanisms for some diseases such as celiac disease (CD). If patients with high levels of anti-tissue transglutaminase (anti-tTG) immunoglobulin A (IgA) antibodies have normal duodenal histology, they are followed as potential CD patients. The aim of this study was to investigate the effect of synbiotic use on the blood levels of anti-tTG antibodies in children. METHODS Eighty-two patients with high anti-tTG levels were included in this study. Patients were randomly divided into two groups. The synbiotic group was treated with a daily dose of a synbiotic including multi-strain probiotics for 20 days. The control group was not administered any medication. Anti-tTG values at baseline and repeat measurements and the percentage change in anti-tTG levels between groups were compared. RESULTS The anti-tTG level at baseline was 36 U/mL (interquartile range [IQR], 26.4-68 U/mL) in the synbiotic group, and it decreased significantly to 13 U/mL (IQR, 6.5-27.5 U/mL) after 20 days (p<0.05). The anti-tTG level at baseline was 46 U/mL (IQR, 31-89 U/mL) in the control group, which also decreased significantly to 23 U/mL (IQR, 7-41 U/mL) after 20 days (p<0.05). Anti-tTG levels exhibited 73% and 56% decreases in the synbiotic and control groups, respectively (p<0.05). CONCLUSION It may be speculated that a synbiotic supplementation can contribute to decreasing anti-tTG levels in children with potential CD.
Collapse
Affiliation(s)
- Kaan Demiroren
- Department of Pediatric Gastroenterology, University of Health Sciences, Yuksek Ihtisas Teaching Hospital, Bursa, Turkey
| |
Collapse
|
32
|
Peng M, Tabashsum Z, Anderson M, Truong A, Houser AK, Padilla J, Akmel A, Bhatti J, Rahaman SO, Biswas D. Effectiveness of probiotics, prebiotics, and prebiotic-like components in common functional foods. Compr Rev Food Sci Food Saf 2020; 19:1908-1933. [PMID: 33337097 DOI: 10.1111/1541-4337.12565] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
The bioactive ingredients in commonly consumed foods include, but are not limited to, prebiotics, prebiotic-like components, probiotics, and postbiotics. The bioactive ingredients in functional foods have also been associated with beneficial effects on human health. For example, they aid in shaping of gut microflora and promotion of immunity. These functional components also contribute in preventing serious diseases such as cardiovascular malfunction and tumorigenesis. However, the specific mechanisms of these positive influences on human health are still under investigation. In this review, we aim to emphasize the major contents of probiotics, prebiotics, and prebiotic-like components commonly found in consumable functional foods, and we present an overview of direct and indirect benefits they provide on human health. The major contributors are certain families of metabolites, specifically short-chain fatty acids and polyunsaturated fatty acids produced by probiotics, and prebiotics, or prebiotic-like components such as flavonoids, polyphenols, and vitamins that are found in functional foods. These functional ingredients in foods influence the gut microbiota by stimulating the growth of beneficial microbes and the production of beneficial metabolites that, in turn, have direct benefits to the host, while also providing protection from pathogens and maintaining a balanced gut ecosystem. The complex interactions that arise among functional food ingredients, human physiology, the gut microbiota, and their respective metabolic pathways have been found to minimize several factors that contribute to the incidence of chronic disease, such as inflammation oxidative stress.
Collapse
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Zajeba Tabashsum
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Mary Anderson
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Andy Truong
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Ashley K Houser
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Joselyn Padilla
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Ahlam Akmel
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Jacob Bhatti
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Sciences, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland.,Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland.,Center for Food Safety and Security Systems, University of Maryland, College Park, Maryland
| |
Collapse
|
33
|
Saeedi BJ, Liu KH, Owens JA, Hunter-Chang S, Camacho MC, Eboka RU, Chandrasekharan B, Baker NF, Darby TM, Robinson BS, Jones RM, Jones DP, Neish AS. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell Metab 2020; 31:956-968.e5. [PMID: 32213347 PMCID: PMC7329068 DOI: 10.1016/j.cmet.2020.03.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/09/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
Many studies have suggested a role for gut-resident microbes (the "gut microbiome") in modulating host health; however, the mechanisms by which they impact systemic physiology remain largely unknown. In this study, metabolomic and transcriptional profiling of germ-free and conventionalized mouse liver revealed an upregulation of the Nrf2 antioxidant and xenobiotic response in microbiome-replete animals. Using a Drosophila-based screening assay, we identified members of the genus Lactobacillus capable of stimulating Nrf2. Indeed, the human commensal Lactobacillus rhamnosus GG (LGG) potently activated Nrf2 in the Drosophila liver analog and the murine liver. This activation was sufficient to protect against two models of oxidative liver injury, acetaminophen overdose and acute ethanol toxicity. Characterization of the portal circulation of LGG-treated mice by tandem mass spectrometry identified a small molecule activator of Nrf2, 5-methoxyindoleacetic acid, produced by LGG. Taken together, these data demonstrate a mechanism by which intestinal microbes modulate hepatic susceptibility to oxidative injury.
Collapse
Affiliation(s)
- Bejan J Saeedi
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ken H Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joshua A Owens
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sarah Hunter-Chang
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mary C Camacho
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Richard U Eboka
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bindu Chandrasekharan
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nusaiba F Baker
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Trevor M Darby
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brian S Robinson
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rheinallt M Jones
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Andrew S Neish
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
34
|
Orlando A, Chimienti G, Lezza AMS, Pesce V, Gigante I, D’Attoma B, Russo F. Lactobacillus Rhamnosus GG Affects the BDNF System in Brain Samples of Wistar Rats with Pepsin-Trypsin-Digested Gliadin (PTG)-Induced Enteropathy. Nutrients 2020; 12:nu12030629. [PMID: 32120967 PMCID: PMC7146293 DOI: 10.3390/nu12030629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
Celiac disease (CD) presents as chronic low-grade inflammation of the small intestine often characterized by psychiatric comorbidities. The brain-derived neurotrophic factor (BDNF), which we have shown to be reduced in the serum of CD patients, acts as the bridge between immune activation and the nervous system adaptive response. Since Lactobacillus has been shown to upregulate BDNF, this study aimed to evaluate whether the administration of Lactobacillus rhamnosus GG (L.GG) could positively affect the brain BDNF system in rats mimicking the CD lesions. Data have shown that the administration of pepsin-trypsin digested gliadin (PTG) and L.GG alter the levels of mature BDNF (mBDNF), as evaluated by Western blotting. PTG provoked a reduction of mBDNF compared to controls, and a compensatory increase of its receptor TrkB. L.GG induced a slight positive effect on mBDNF levels under normal conditions, while it was able to rescue the PTG-induced reduced expression of mBDNF. The curative effect of L.GG was finely tuned, accompanied by the reduction of TrkB, probably to avoid the effect of excessive BDNF.
Collapse
Affiliation(s)
- Antonella Orlando
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology “S. de Bellis” Research Hospital, 70013 Castellana Grotte (Ba), Italy; (A.O.); (B.D.)
| | - Guglielmina Chimienti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70100 Bari, Italy; (G.C.); (A.M.S.L.); (V.P.)
| | - Angela Maria Serena Lezza
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70100 Bari, Italy; (G.C.); (A.M.S.L.); (V.P.)
| | - Vito Pesce
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70100 Bari, Italy; (G.C.); (A.M.S.L.); (V.P.)
| | - Isabella Gigante
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology “S. de Bellis” Research Hospital, 70013 Castellana Grotte (Ba), Italy;
| | - Benedetta D’Attoma
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology “S. de Bellis” Research Hospital, 70013 Castellana Grotte (Ba), Italy; (A.O.); (B.D.)
| | - Francesco Russo
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology “S. de Bellis” Research Hospital, 70013 Castellana Grotte (Ba), Italy; (A.O.); (B.D.)
- Correspondence: ; Tel.: +3908-0499-4129
| |
Collapse
|
35
|
Penny HA, Baggus EMR, Rej A, Snowden JA, Sanders DS. Non-Responsive Coeliac Disease: A Comprehensive Review from the NHS England National Centre for Refractory Coeliac Disease. Nutrients 2020; 12:E216. [PMID: 31947666 PMCID: PMC7019917 DOI: 10.3390/nu12010216] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Coeliac disease is a common small intestinal enteropathy which manifests following ingestion of gluten in genetically susceptible individuals. Since gluten was identified as the driving factor in coeliac disease, the gluten-free diet (GFD) has remained the mainstay of treatment. While most individuals will display improvement in symptoms and signs of coeliac disease following institution of the GFD, up to 30% will continue to experience symptoms and/or have persisting intestinal inflammation. These individuals can be classified as having non-responsive coeliac disease (NRCD), which may be associated with dietary indiscretion, slow healing, refractory coeliac disease, and/or an alternative condition. The purpose of this review is to provide an overview of the causes of NRCD in adults, highlight a systematic approach to investigate these patients, and appraise the latest management aspects of this subset of coeliac disease.
Collapse
Affiliation(s)
- Hugo A. Penny
- Academic Unit of Gastroenterology, University of Sheffield, Sheffield S10 2TN, UK; (H.A.P.); (E.M.R.B.); (A.R.)
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester M13 9PL, UK
| | - Elisabeth M. R. Baggus
- Academic Unit of Gastroenterology, University of Sheffield, Sheffield S10 2TN, UK; (H.A.P.); (E.M.R.B.); (A.R.)
| | - Anupam Rej
- Academic Unit of Gastroenterology, University of Sheffield, Sheffield S10 2TN, UK; (H.A.P.); (E.M.R.B.); (A.R.)
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK;
| | - David S. Sanders
- Academic Unit of Gastroenterology, University of Sheffield, Sheffield S10 2TN, UK; (H.A.P.); (E.M.R.B.); (A.R.)
| |
Collapse
|
36
|
Renga G, Moretti S, Oikonomou V, Borghi M, Zelante T, Paolicelli G, Costantini C, De Zuani M, Villella VR, Raia V, Del Sordo R, Bartoli A, Baldoni M, Renauld JC, Sidoni A, Garaci E, Maiuri L, Pucillo C, Romani L. IL-9 and Mast Cells Are Key Players of Candida albicans Commensalism and Pathogenesis in the Gut. Cell Rep 2019; 23:1767-1778. [PMID: 29742432 PMCID: PMC5976578 DOI: 10.1016/j.celrep.2018.04.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 02/14/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022] Open
Abstract
Candida albicans is implicated in intestinal diseases. Identifying host signatures that discriminate between the pathogenic versus commensal nature of this human commensal is clinically relevant. In the present study, we identify IL-9 and mast cells (MCs) as key players of Candida commensalism and pathogenicity. By inducing TGF-β in stromal MCs, IL-9 pivotally contributes to mucosal immune tolerance via the indoleamine 2,3-dioxygenase enzyme. However, Candida-driven IL-9 and mucosal MCs also contribute to barrier function loss, dissemination, and inflammation in experimental leaky gut models and are upregulated in patients with celiac disease. Inflammatory dysbiosis occurs with IL-9 and MC deficiency, indicating that the activity of IL-9 and MCs may go beyond host immunity to include regulation of the microbiota. Thus, the output of the IL-9/MC axis is highly contextual during Candida colonization and reveals how host immunity and the microbiota finely tune Candida behavior in the gut. IL-9/IL-9R signaling affects MC function in mucosal candidiasis IL-9 and mucosal MCs contribute to barrier function loss in leaky gut models IL-9 and stromal MCs induce local protective tolerance in infection via IDO1 IL-9 and mucosal MCs expand and IDO1 decreases in human celiac disease
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Giuseppe Paolicelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Marco De Zuani
- Department of Medical and Biological Science, University of Udine, 33100 Udine, Italy
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, 80131 Naples, Italy
| | - Rachele Del Sordo
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Monia Baldoni
- Department of Medicine, University of Perugia, 06132 Perugia, Italy
| | | | - Angelo Sidoni
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Enrico Garaci
- San Raffaele Pisana, IRCCS, Telematic University and University of Tor Vergata, 00163 Rome, Italy
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Carlo Pucillo
- Department of Medical and Biological Science, University of Udine, 33100 Udine, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
37
|
Guo JW, Wu XN, Cheng RY, Shen X, Cheng G, Yu LX, Li M, He F. Oral administration of vancomycin to neonatal mice could alter their immunity and allergic sensibility late in adulthood. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2019; 38:129-139. [PMID: 31763116 PMCID: PMC6856516 DOI: 10.12938/bmfh.19-008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 11/28/2022]
Abstract
The prevalence of allergy has increased over the past decades, and this may be attributed in part to the intestinal microbiota dysfunction caused by antibiotics during early life. In this
study, we evaluated how vancomycin could impair the intestinal microbiota during early life and then, consequently, alter susceptibilities to allergic diseases and related immunity in late
adulthood. BALB/c (n=54) neonatal mice were used in this study. Mice in the vancomycin group were orally administered vancomycin for 21 days, while those in the allergy and control groups
were perfused with the same volume of saline solution. Then, mice in the allergy and vancomycin groups were immunized with intraperitoneal ovalbumin with alum. At postnatal day 21,
vancomycin significantly alter the fecal microbiota, with lower Bacteroidetes and Firmicutes counts and higher Proteobacteria counts. At
postnatal day 56, the Bacteroidetes count was still significantly lower in vancomycin-treated mice. The serum IgE level in the control group was significantly lower than
that in the vancomycin and allergy groups. The serum interleukin (IL)-6 level and the IL-4/interferon (IFN)-γ values were significantly higher in the vancomycin-treated mice, but the serum
IL-17A level was lower than that in the control group. These results indicate that modifications of the intestinal microbiota composition during early life may be, at least in part, the key
mechanism underlying the effect of vancomycin that influences the immune function of host animals in the adult stages.
Collapse
Affiliation(s)
- Jia Wen Guo
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Xiao Na Wu
- West China Second Hospital, Sichuan University/West China Women's and Children's Hospital, 610041 Chengdu, Sichuan, China
| | - Ru Yue Cheng
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Xi Shen
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Guo Cheng
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Lan Xiu Yu
- Green's Bioengineering (Shenzhen) Co., Ltd., 518105 Shenzhen, Guangdong, China
| | - Ming Li
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Fang He
- West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, 610041 Chengdu, Sichuan, China
| |
Collapse
|
38
|
Montalban-Arques A, Scharl M. Intestinal microbiota and colorectal carcinoma: Implications for pathogenesis, diagnosis, and therapy. EBioMedicine 2019; 48:648-655. [PMID: 31631043 PMCID: PMC6838386 DOI: 10.1016/j.ebiom.2019.09.050] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/06/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers and leading cause of cancer-related deaths worldwide. In recent years, there has been a growing realisation that lifestyle plays a major role for CRC development and that intestinal microbiota, which are shaped by lifestyle and nutrition habits, may be critically involved in the pathogenesis of CRC. Although the precise mechanisms for how the microbiota contribute to CRC development and progression remain elusive, increasing evidence suggests a direct causative role for the intestinal microbiota in modulating signalling pathways, anti-tumour immune responses and cell proliferation. Recent advances in understanding host-microbe interactions have shed light onto the putative use of intestinal microbiota as a powerful tool in CRC diagnosis and therapy. Here, we will discuss the role of the intestinal microbiota in CRC pathogenesis, their potential utility as diagnostic markers, and consider how microbes could be used in therapeutic approaches for the treatment of CRC.
Collapse
Affiliation(s)
- Ana Montalban-Arques
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
39
|
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: 10.8] [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
|
40
|
Abstract
PURPOSE OF REVIEW Celiac disease (CD) is an autoimmune enteropathy triggered by gluten. The purpose of this review is to examine the major genetic and environmental factors that contribute to CD pathogenesis. RECENT FINDINGS We reviewed the current state of knowledge on the genetic and environmental components that play a role in CD onset. A genome-wide association study (GWAS) analysis has highlighted several genes other than HLA involved in CD. Recent studies have shown that HLA haplotype influences the microbiome composition in infants and that dysbiosis in the intestinal microflora, in turn, contributes to loss of tolerance to gluten. Recently, observational studies have discussed the hypothesis stating that breast-feeding had a protective role against CD onset. CD etiology is influenced by genetic and environmental factors. A better understanding of these components would deepen our knowledge on the mechanisms that lead to loss of tolerance and could help in developing a more "personalized medicine."
Collapse
|
41
|
Labruna G, Nanayakkara M, Pagliuca C, Nunziato M, Iaffaldano L, D'Argenio V, Colicchio R, Budelli AL, Nigro R, Salvatore P, Barone MV, Sacchetti L. Celiac disease-associated Neisseria flavescens decreases mitochondrial respiration in CaCo-2 epithelial cells: Impact of Lactobacillus paracasei CBA L74 on bacterial-induced cellular imbalance. Cell Microbiol 2019; 21:e13035. [PMID: 31042331 PMCID: PMC6618323 DOI: 10.1111/cmi.13035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022]
Abstract
We previously identified a Neisseria flavescens strain in the duodenum of celiac disease (CD) patients that induced immune inflammation in ex vivo duodenal mucosal explants and in CaCo‐2 cells. We also found that vesicular trafficking was delayed after the CD‐immunogenic P31‐43 gliadin peptide‐entered CaCo‐2 cells and that Lactobacillus paracasei CBA L74 (L. paracasei‐CBA) supernatant reduced peptide entry. In this study, we evaluated if metabolism and trafficking was altered in CD‐N. flavescens‐infected CaCo‐2 cells and if any alteration could be mitigated by pretreating cells with L. paracasei‐CBA supernatant, despite the presence of P31‐43. We measured CaCo‐2 bioenergetics by an extracellular flux analyser, N. flavescens and P31‐43 intracellular trafficking by immunofluorescence, cellular stress by TBARS assay, and ATP by bioluminescence. We found that CD‐N. flavescens colocalised more than control N. flavescens with early endocytic vesicles and more escaped autophagy thereby surviving longer in infected cells. P31‐43 increased colocalisation of N. flavescens with early vesicles. Mitochondrial respiration was lower (P < .05) in CD‐N. flavescens‐infected cells versus not‐treated CaCo‐2 cells, whereas pretreatment with L. paracasei‐CBA reduced CD‐N. flavescens viability and improved cell bioenergetics and trafficking. In conclusion, CD‐N. flavescens induces metabolic imbalance in CaCo‐2 cells, and the L. paracasei‐CBA probiotic could be used to correct CD‐associated dysbiosis.
Collapse
Affiliation(s)
- Giuseppe Labruna
- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) SDN, Naples, Italy
| | - Merlin Nanayakkara
- Dipartimento di Scienze Mediche Traslazionali and European Laboratory for the Investigation of Food Induced Disease (ELFID), Università degli Studi di Napoli Federico II, Naples, Italy
| | - Chiara Pagliuca
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Marcella Nunziato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy
| | | | - Valeria D'Argenio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy.,Task Force on Microbiome Studies, Università degli Studi di Napoli Federico II and CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy
| | - Roberta Colicchio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy
| | | | - Roberto Nigro
- Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università di Napoli Federico II, Naples, Italy
| | - Paola Salvatore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Maria Vittoria Barone
- Dipartimento di Scienze Mediche Traslazionali and European Laboratory for the Investigation of Food Induced Disease (ELFID), Università degli Studi di Napoli Federico II, Naples, Italy
| | - Lucia Sacchetti
- CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy.,Task Force on Microbiome Studies, Università degli Studi di Napoli Federico II and CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy
| |
Collapse
|
42
|
Yu LCH. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018; 25:79. [PMID: 30413188 PMCID: PMC6234774 DOI: 10.1186/s12929-018-0483-8] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease which arises as a result of the interaction of genetic, environmental, barrier and microbial factors leading to chronic inflammation in the intestine. Patients with IBD had a higher risk of developing colorectal carcinoma (CRC), of which the subset was classified as colitis-associated cancers. Genetic polymorphism of innate immune receptors had long been considered a major risk factor for IBD, and the mutations were also recently observed in CRC. Altered microbial composition (termed microbiota dybiosis) and dysfunctional gut barrier manifested by epithelial hyperpermeability and high amount of mucosa-associated bacteria were observed in IBD and CRC patients. The findings suggested that aberrant immune responses to penetrating commensal microbes may play key roles in fueling disease progression. Accumulative evidence demonstrated that mucosa-associated bacteria harbored colitogenic and protumoral properties in experimental models, supporting an active role of bacteria as pathobionts (commensal-derived opportunistic pathogens). Nevertheless, the host factors involved in bacterial dysbiosis and conversion mechanisms from lumen-dwelling commensals to mucosal pathobionts remain unclear. Based on the observation of gut leakiness in patients and the evidence of epithelial hyperpermeability prior to the onset of mucosal histopathology in colitic animals, it was postulated that the epithelial barrier dysfunction associated with mucosal enrichment of specific bacterial strains may predispose the shift to disease-associated microbiota. The speculation of leaky gut as an initiating factor for microbiota dysbiosis that eventually led to pathological consequences was proposed as the "common ground hypothesis", which will be highlighted in this review. Overall, the understanding of the core interplay between gut microbiota and epithelial barriers at early subclinical phases will shed light to novel therapeutic strategies to manage chronic inflammatory disorders and colitis-associated cancers.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Suite 1020, #1 Jen-Ai Rd. Sec. 1, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
43
|
Tindall AM, Petersen KS, Lamendella R, Shearer GC, Murray-Kolb LE, Proctor DN, Kris-Etherton PM. Tree Nut Consumption and Adipose Tissue Mass: Mechanisms of Action. Curr Dev Nutr 2018; 2:nzy069. [PMID: 30488045 PMCID: PMC6252345 DOI: 10.1093/cdn/nzy069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/18/2018] [Accepted: 08/02/2018] [Indexed: 01/03/2023] Open
Abstract
There is concern that tree nuts may cause weight gain due to their energy density, yet evidence shows that tree nuts do not adversely affect weight status. Epidemiologic and experimental studies have shown a reduced risk of chronic diseases with tree nut consumption without an increased risk of weight gain. In fact, tree nuts may protect against weight gain and benefit weight-loss interventions. However, the relation between tree nut consumption and adiposity is not well understood at the mechanistic level. This review summarizes the proposed underlying mechanisms that might account for this relation. Evidence suggests that tree nuts may affect adiposity through appetite control, displacement of unfavorable nutrients, increased diet-induced thermogenesis, availability of metabolizable energy, antiobesity action of bioactive compounds, and improved functionality of the gut microbiome. The gut microbiome is a common factor among these mechanisms and may mediate, in part, the relation between tree nut consumption and reduced adiposity. Further research is needed to understand the impact of tree nuts on the gut microbiome and how the gut microbial environment affects the nutrient absorption and metabolism of tree nuts. The evidence to date suggests that tree nut consumption favorably affects body composition through different mechanisms that involve the gut microbiome. A better understanding of these mechanisms will contribute to the evolving science base that addresses the causes and treatments for overweight and obesity.
Collapse
Affiliation(s)
- Alyssa M Tindall
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Kristina S Petersen
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | | | - Gregory C Shearer
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Laura E Murray-Kolb
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - David N Proctor
- Departments of Kinesiology, The Pennsylvania State University, University Park, PA
| | - Penny M Kris-Etherton
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
44
|
Clinical intervention using Bifidobacterium strains in celiac disease children reveals novel microbial modulators of TNF-α and short-chain fatty acids. Clin Nutr 2018; 38:1373-1381. [PMID: 29960810 DOI: 10.1016/j.clnu.2018.06.931] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/30/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Celiac disease (CD) is an immune-mediated systemic disease, caused by ingestion of gluten in genetically predisposed individuals. Gut microbiota dysbiosis might play a significant role in pathogenesis of chronic enteropathies and its modulation can be used as an intervention strategy in CD as well. In this study, we aimed to identify correlations between fecal microbiota, serum tumor necrosis factor alpha (TNF-α) and fecal short-chain fatty acids (SCFAs) in healthy children and children with CD after administration of probiotic Bifidobacterium breve BR03 and B632. METHODS A double-blind placebo-controlled study enrolled 40 children with CD (CD) and 16 healthy children (HC). CD children were randomly allocated into two groups, of which 20 belonged to the placebo (PL) group and 20 to the Probiotic (PR) group. The PR group received a probiotic formulation containing a mixture of 2 strains, B. breve BR03 (DSM 16604) and B. breve B632 (DSM 24706) in 1:1 ratio for 3 months. Subsequently, for statistical analysis, blood and fecal samples from CD children (on enrolment - T0 and after 3 months, at the end of intervention with probiotic/placebo - T1) and HC children were used. The HC group was sampled only once (T0). RESULTS Verrucomicrobia, Parcubacteria and some yet unknown phyla of Bacteria and Archaea may be involved in the disease, indicated by a strong correlation to TNF-α. Likewise, Proteobacteria strongly correlated with fecal SCFAs concentration. The effect of probiotic administration has disclosed a negative correlation between Verrucomicrobia, some unknown phyla of Bacteria, Synergistetes, Euryarchaeota and some SCFAs, turning them into an important target in microbiome restoration process. Synergistetes and Euryarchaeota may have a role in the anti-inflammatory process in healthy human gut. CONCLUSIONS Our results highlight new phyla, which may have an important relation to disease-related parameters, CD itself and health.
Collapse
|
45
|
Nobel YR, Axelrad J, Lewis SK, Whittier S, Lawlor G, Lichtiger S, Green PHR, Lebwohl B. Stool PCR for Gastrointestinal Pathogens in Patients With and Without Immune-Mediated Intestinal Diseases. Dig Dis Sci 2018; 63:996-1002. [PMID: 29411208 DOI: 10.1007/s10620-018-4959-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients with celiac disease and inflammatory bowel disease, two immune-mediated luminal conditions, have higher rates of certain infections than healthy counterparts. The prevalence of many gastrointestinal infections in these patients, however, is unknown. AIMS Using a novel clinical stool pathogen PCR test, we investigated the hypothesis that patients with celiac disease/inflammatory bowel disease had different distributions of diarrheal pathogens than other patients. METHODS We performed a retrospective cohort study of outpatients who underwent stool pathogen testing with the FilmArray Gastrointestinal PCR Panel (BioFire Diagnostics, Salt Lake City, UT) at our institution from January 1 to December 31, 2015. Rates of pathogens were measured in patients with or without celiac disease/inflammatory bowel disease. RESULTS Of 955 patients, 337 had positive test for any pathogen, with 465 bacterial, parasitic, or viral pathogens identified. One hundred and twenty-seven patients (13.3%) had celiac disease or inflammatory bowel disease, of which 29/127 (22.8%) had a positive test, compared to 308/828 other patients (37.2%) (p = 0.002). Patients with celiac disease/inflammatory bowel disease had significantly fewer viruses (1.6 vs. 8.1% of patients; p = 0.008) and parasites (0 vs. 3.3%; p = 0.039), with nonsignificant trend toward fewer bacteria (21.3 vs. 29.2%; p = 0.063). Escherichia coli species were most common in both populations. CONCLUSIONS Stool PCR identified numerous pathogens in patients with or without celiac disease/inflammatory bowel disease. Patients with celiac disease/inflammatory bowel disease were significantly less likely to have any pathogen identified, and had significantly fewer viruses and parasites. In this population, knowledge of common pathogens can guide diagnostic evaluation and offer opportunities for treatment.
Collapse
Affiliation(s)
- Yael R Nobel
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Jordan Axelrad
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Suzanne K Lewis
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Susan Whittier
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Garrett Lawlor
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Simon Lichtiger
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Peter H R Green
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Benjamin Lebwohl
- Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, USA.
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
- The Celiac Disease Center at Columbia University, 180 Fort Washington Avenue, Suite 936, New York, NY, 10032, USA.
| |
Collapse
|
46
|
Erturk-Hasdemir D, Kasper DL. Finding a needle in a haystack: Bacteroides fragilis polysaccharide A as the archetypical symbiosis factor. Ann N Y Acad Sci 2018. [PMID: 29528123 DOI: 10.1111/nyas.13660] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Starting from birth, all animals develop a symbiotic relationship with their resident microorganisms that benefits both the microbe and the host. Recent advances in technology have substantially improved our ability to direct research toward the identification of important microbial species that affect host physiology. The identification of specific commensal molecules from these microbes and their mechanisms of action is still in its early stages. Polysaccharide A (PSA) of Bacteroides fragilis is the archetypical example of a commensal molecule that can modulate the host immune system in health and disease. This zwitterionic polysaccharide has a critical impact on the development of the mammalian immune system and also on the stimulation of interleukin 10-producing CD4+ T cells; consequently, PSA confers benefits to the host with regard to experimental autoimmune, inflammatory, and infectious diseases. In this review, we summarize the current understanding of the immunomodulatory effects of B. fragilis PSA and discuss these effects as a novel immunological paradigm. In particular, we discuss recent advances in our understanding of the unique functional mechanisms of this molecule and its therapeutic potential, and we review the recent literature in the field of microbiome research aimed at discovering new commensal products and their immunomodulatory potential.
Collapse
Affiliation(s)
- Deniz Erturk-Hasdemir
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Dennis L Kasper
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Immunopathology of childhood celiac disease-Key role of intestinal epithelial cells. PLoS One 2017; 12:e0185025. [PMID: 28934294 PMCID: PMC5608296 DOI: 10.1371/journal.pone.0185025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/04/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND & AIMS Celiac disease is a chronic inflammatory disease of the small intestine mucosa due to permanent intolerance to dietary gluten. The aim was to elucidate the role of small intestinal epithelial cells in the immunopathology of celiac disease in particular the influence of celiac disease-associated bacteria. METHODS Duodenal biopsies were collected from children with active celiac disease, treated celiac disease, and clinical controls. Intestinal epithelial cells were purified and analyzed for gene expression changes at the mRNA and protein levels. Two in vitro models for human intestinal epithelium, small intestinal enteroids and polarized tight monolayers, were utilized to assess how interferon-γ, interleukin-17A, celiac disease-associated bacteria and gluten influence intestinal epithelial cells. RESULTS More than 25 defense-related genes, including IRF1, SPINK4, ITLN1, OAS2, CIITA, HLA-DMB, HLA-DOB, PSMB9, TAP1, BTN3A1, and CX3CL1, were significantly upregulated in intestinal epithelial cells at active celiac disease. Of these genes, 70% were upregulated by interferon-γ via the IRF1 pathway. Most interestingly, IRF1 was also upregulated by celiac disease-associated bacteria. The NLRP6/8 inflammasome yielding CASP1 and biologically active interleukin-18, which induces interferon-γ in intraepithelial lymphocytes, was expressed in intestinal epithelial cells. CONCLUSION A key factor in the epithelial reaction in celiac disease appears to be over-expression of IRF1 that could be inherent and/or due to presence of undesirable microbes that act directly on IRF1. Dual activation of IRF1 and IRF1-regulated genes, both directly and via the interleukin-18 dependent inflammasome would drastically enhance the inflammatory response and lead to the pathological situation seen in active celiac disease.
Collapse
|