1
|
San-Martin MI, Chamizo-Ampudia A, Sanchiz Á, Ferrero MÁ, Martínez-Blanco H, Rodríguez-Aparicio LB, Navasa N. Microbiome Markers in Gastrointestinal Disorders: Inflammatory Bowel Disease, Colorectal Cancer, and Celiac Disease. Int J Mol Sci 2025; 26:4818. [PMID: 40429958 PMCID: PMC12112578 DOI: 10.3390/ijms26104818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/06/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Intestinal microbiota and the host's immune system form a symbiotic alliance that sustains normal development and function in the human gut. Changes such as dietary habits among societies in developed countries have led to the development of unbalanced microbial populations in the gut, likely contributing to the dramatic increase in inflammatory diseases in the last few decades. Recent advances in DNA sequencing technologies have tremendously helped to characterize the microbiome associated with disease, both in identifying global alterations and discovering specific biomarkers that potentially contribute to disease pathogenesis, as evidenced by animal studies. Beyond bacterial alterations, non-bacterial components such as fungi, viruses, and microbial metabolites have been implicated in these diseases, influencing immune responses and gut homeostasis. Multi-omics approaches integrating metagenomics, metabolomics, and transcriptomics offer a more comprehensive understanding of the microbiome's role in disease pathogenesis, paving the way for innovative diagnostic and therapeutic strategies. Unraveling the metagenomic profiles associated with disease may facilitate earlier diagnosis and intervention, as well as the development of more personalized and effective therapeutic strategies. This review synthesizes recent and relevant microbiome research studies aimed at characterizing the microbial signatures associated with inflammatory bowel disease, colorectal cancer, and celiac disease.
Collapse
Affiliation(s)
- M. Isabel San-Martin
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Alejandro Chamizo-Ampudia
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - África Sanchiz
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Miguel Ángel Ferrero
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Honorina Martínez-Blanco
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Leandro Benito Rodríguez-Aparicio
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Nicolás Navasa
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain; (M.I.S.-M.); (A.C.-A.); (M.Á.F.); (H.M.-B.); (N.N.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| |
Collapse
|
2
|
Leo S, Leonard MM, Valitutti F, Fasano A. Gut dysbiosis: cause or consequence of intestinal inflammation in celiac disease? Expert Rev Gastroenterol Hepatol 2025; 19:505-513. [PMID: 40133841 DOI: 10.1080/17474124.2025.2483406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION Celiac disease (CeD) is an immune-mediated condition that occurs in genetically predisposed individuals ingesting gluten. It is characterized by enteropathy leading to both gastrointestinal and extra-intestinal symptoms. The prevalence of CeD has increased world-wide. Evidence suggests that genetic predisposition and exposure to gluten are necessary but not sufficient for CeD onset, implying that other unknown factors are at play in its pathogenesis. AREAS COVERED This review summarizes the current knowledge on the contribution of the gut microbiota to CeD pathogenesis, aiming to address the question of whether it is the cause or consequence of the celiac enteropathy. We reviewed the current literature (studies published in PubMed database between 2007 and 2023), linking gut microbiota dysbiosis and CeD, focusing specifically on prospective birth cohorts' studies and discussing how multi-omics and artificial intelligence (AI) could transform the diagnosis of CeD in a personalized medicine approach. EXPERT OPINION A multi-omic approach will allow for better clarification of the pivotal role of the microbiome in epigenetically triggering CeD pathogenesis. Further, the combination of multi-omics results with AI would pave the way to an improved CeD diagnosis and to the identification of new personalized therapeutic interventions.
Collapse
Affiliation(s)
- Stefano Leo
- European Biomedical Research Institute of Salerno, Salerno, Italy
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
| | - Francesco Valitutti
- Pediatrics Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno, Salerno, Italy
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Kelley K, Dogru D, Huang Q, Yang Y, Palm NW, Altindis E, Ludvigsson J. Children who develop celiac disease are predicted to exhibit distinct metabolic pathways among their gut microbiota years before diagnosis. Microbiol Spectr 2025; 13:e0146824. [PMID: 39902908 PMCID: PMC11878042 DOI: 10.1128/spectrum.01468-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Celiac disease (CD) is an autoimmune disease caused by a loss of gluten tolerance in genetically predisposed individuals. While 30%-40% of people possess the predisposing alleles, only 1%-2% are diagnosed with CD, suggesting that environmental factors are involved in disease pathogenesis. To determine an association between pediatric CD and the gut microbiome, we analyzed fecal samples from a prospective cohort study (ABIS). These samples were collected from children who later developed CD (CD progressors) and age-matched healthy children (at ages 1, 2.5, and 5) with similar HLA genotypes, breastfeeding durations, and gluten exposure times. We previously reported gut microbiome differences at ages 2.5 and 5 in this cohort; here, we present findings from samples collected at age 1 (n = 5). We identified 14 ASVs differing significantly between CD progressors and controls, including taxa linked to CD pathogenesis. CD progressors had increased Firmicutes and higher alpha diversity in IgA- bacteria. Using PICRUSt, we analyzed metabolic pathways enriched in CD progressors compared to controls at ages 1, 2.5, and 5 (n = 5-16), revealing enriched inflammatory and pathogenic pathways potentially contributing to CD-related immune dysregulation. While results are based on the primary EdgeR analysis, we also applied a non-parametric method of statistical analysis, reporting those results with supplementary figures. In conclusion, our findings suggest distinct metabolic pathways enriched in the gut microbiome of CD progressors years before diagnosis, which could inform targeted therapeutics for CD. As discussed in the limitations section, this small pilot study should be replicated with larger sample sizes for broader generalization. IMPORTANCE We analyzed gut microbiome data from children who later developed celiac disease (CD progressors) compared to healthy children in the first 5 years of life. Using fecal samples corresponding to the three phases of gut microbiome development, we uncovered enriched functional microbial pathways in CD progressors at age 1. Some of these pathways, implicated in bacterial pathogenesis, microbiota modulation, and inflammation, have been correlated with CD. We also identified taxa in CD progressors at age 1 including Lachnospiraceae, Alistipes, and Bifidobacterium dentium that were previously associated with CD. These findings suggest a potential role for these taxa and enriched pathways in pediatric CD onset years before diagnosis, highlighting potential for early interventions. While the findings of this exploratory study should be validated with larger sample sizes, our study suggests microbial metabolic pathways related to CD onset, enhancing our understanding of CD pathogenesis and the role of gut microbiome-mediated early alterations.
Collapse
Affiliation(s)
- Kristina Kelley
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Dogus Dogru
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Qian Huang
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Johnny Ludvigsson
- Crown Princess Victoria’s Children’s Hospital, Region Östergötland, Linköping, Sweden
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Simpson HL, Smits E, Moerkens R, Wijmenga C, Mooiweer J, Jonkers IH, Withoff S. Human organoids and organ-on-chips in coeliac disease research. Trends Mol Med 2025; 31:117-137. [PMID: 39448329 DOI: 10.1016/j.molmed.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Coeliac disease (CeD) is an immune-mediated disorder characterised by gluten-triggered inflammation and damage in the small intestine, with lifelong gluten-free diet (GFD) as the only treatment. It is a multifactorial disease, involving genetic and environmental susceptibility factors, and its complexity and lack of comprehensive human model systems have hindered understanding of its pathogenesis and development of new treatments. Therefore, it is crucial to establish systems that recapitulate patient genetic background and the interactions between the small intestinal epithelial barrier, immune cells, and environment that contribute to CeD. In this review, we discuss disease complexity, recent advances in stem cell biology, organoids, tissue co-cultures, and organ-on-chip (OoC) systems that facilitate the development of comprehensive human model systems, and model applications in preclinical studies of potential treatments.
Collapse
Affiliation(s)
- Hanna L Simpson
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Eline Smits
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Renée Moerkens
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Joram Mooiweer
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Iris H Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
5
|
Fundora MP, Calamaro CJ, Wu Y, Brown AM, St John A, Keiffer R, Xiang Y, Liu K, Gillespie S, Denning PW, Sanders-Lewis K, Seitter B, Bai J. Microbiome and Growth in Infants with Congenital Heart Disease. J Pediatr 2024; 274:114169. [PMID: 38944188 DOI: 10.1016/j.jpeds.2024.114169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVE To profile the gut microbiome (GM) in infants with congenital heart disease (CHD) undergoing cardiac surgery compared with matched infants and to investigate the association with growth (weight, length, and head circumference). STUDY DESIGN A prospective study in the cardiac intensive care unit at Children's Healthcare of Atlanta and newborn nursery within the Emory Healthcare system. Characteristics including weight, length, head circumference, and surgical variables were collected. Fecal samples were collected presurgery (T1), postsurgery (T2), and before discharge (T3), and once for controls. 16 small ribosomal RNA subunit V4 gene was sequenced from fecal samples and classified into taxonomy using Silva v138. RESULTS There were 34 children with CHD (cases) and 34 controls. Cases had higher alpha-diversity, and beta-diversity showed significant dissimilarities compared with controls. GM was associated with lower weight and smaller head circumference (z-score < 2). Lower weight was associated with less Acinetobacter, Clostridioides, Parabacteroides, and Escherichia-Shigella. Smaller head circumference with more Veillonella, less Acinetobacter, and less Parabacteroides. CONCLUSIONS Significant differences in GM diversity and abundance were observed between infants with CHD and control infants. Lower weight and smaller head circumference were associated with distinct GM patterns. Further study is needed to understand the longitudinal effect of microbial dysbiosis on growth in children with CHD.
Collapse
Affiliation(s)
- Michael P Fundora
- Children's Healthcare of Atlanta Cardiology, Emory School of Medicine, Emory University, Atlanta, GA
| | - Christina J Calamaro
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA; Children's Healthcare of Atlanta, Heart Center, Atlanta, GA
| | - Yuhua Wu
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Ann-Marie Brown
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA; Children's Healthcare of Atlanta, Heart Center, Atlanta, GA; ECU Health, Greenville, NC
| | - Amelia St John
- Children's Healthcare of Atlanta, Heart Center, Atlanta, GA
| | | | - Yijin Xiang
- Children's Healthcare of Atlanta, Emory University School of Medicine, Biostatistics, Atlanta, GA
| | - Katie Liu
- Children's Healthcare of Atlanta, Emory University School of Medicine, Biostatistics, Atlanta, GA
| | - Scott Gillespie
- Children's Healthcare of Atlanta, Emory University School of Medicine, Biostatistics, Atlanta, GA
| | - Patricia Wei Denning
- Children's Healthcare of Atlanta, Neonatology, Emory University School of Medicine, Atlanta, GA
| | - Kolby Sanders-Lewis
- Children's Healthcare of Atlanta, Research Core, Emory University School of Medicine, Atlanta, GA
| | - Brooke Seitter
- Children's Healthcare of Atlanta, Research Core, Emory University School of Medicine, Atlanta, GA
| | - Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA.
| |
Collapse
|
6
|
Stanciu D, Staykov H, Dragomanova S, Tancheva L, Pop RS, Ielciu I, Crișan G. Gluten Unraveled: Latest Insights on Terminology, Diagnosis, Pathophysiology, Dietary Strategies, and Intestinal Microbiota Modulations-A Decade in Review. Nutrients 2024; 16:3636. [PMID: 39519469 PMCID: PMC11547711 DOI: 10.3390/nu16213636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A decade of research on gluten-related disorders (GRDs) is reviewed in this study, with a particular emphasis on celiac disease (CD) and non-celiac gluten sensitivity (NCGS). GRDs are triggered by the ingestion of gluten and gluten-like proteins found in wheat, barley, and rye. These proteins lead to intestinal damage in celiac disease, an autoimmune condition characterized by villous atrophy and a variety of gastrointestinal and extraintestinal symptoms. More enigmatic and less understood, NCGS involves symptoms similar to CD but without the immunological reaction or intestinal damage. Recent years have seen advances in the understanding of GRDs, particularly in connection to how intestinal microbiota influences disease progression and patient outcomes. The gluten-free diet (GFD) is still the standard therapy recommended for GRDs despite significant challenges, as discussed in this article. Precise diagnostic methods, patient education and dietary counseling are critical for improving patients' quality of life. The purpose of this review is to provide a more clear and up-to-date understanding of GRDs, and to help further research on this important topic.
Collapse
Affiliation(s)
- Dana Stanciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Hristian Staykov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Radu Samuel Pop
- 3rd Department of Pediatrics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania;
| | - Irina Ielciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Gianina Crișan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| |
Collapse
|
7
|
Roque A, Zanker J, Brígido S, Tomaz MB, Gonçalves A, Barbeiro S, Benítez-Páez A, Pereira SG. Dietary patterns drive loss of fiber-foraging species in the celiac disease patients gut microbiota compared to first-degree relatives. Gut Pathog 2024; 16:58. [PMID: 39375796 PMCID: PMC11459851 DOI: 10.1186/s13099-024-00643-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Celiac disease is an autoimmune disorder triggered by dietary gluten in genetically predisposed individuals that primarily affects the small intestine. Studies have reported differentially abundant bacterial taxa in the gut microbiota of celiac patients compared with non-celiac controls. However, findings across studies have inconsistencies and no microbial signature of celiac disease has been defined so far. RESULTS Here, we showed, by comparing celiac patients with their non-celiac 1st-degree relatives, that bacterial communities of related individuals have similar species occurrence and abundance compared with non-relatives, regardless the disease status. We also found in celiac patients a loss of bacterial species associated with fiber degradation, and host metabolic and immune modulation, as ruminiclostridia, ruminococci, Prevotella, and Akkermansia muciniphila species. We demonstrated that the differential abundance of bacterial species correlates to different dietary patterns observed between the two groups. For instance, Ruminiclostridium siraeum, Ruminococcus bicirculans, and Bacteroides plebeious, recognized as fiber-degraders, appear more abundant in non-celiac 1st-degree relatives, which have a vegetable consumption pattern higher than celiac patients. Pattern of servings per day also suggests a possible link between these species' abundance and daily calorie intake. CONCLUSIONS Overall, we evidenced that a kinship approach could be valuable in unveiling potential celiac disease microbial traits, as well as the significance of dietary factors in shaping microbial profiles and their influence on disease development and progression. Our results pave the way for designing and adopting novel dietary strategies based on gluten-free fiber-enriched ingredients to improve disease management and patients' quality of life.
Collapse
Affiliation(s)
- Ana Roque
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal
| | - Joyce Zanker
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal
| | - Sara Brígido
- Labeto, Centro de Análises Bioquímicas SA, Beatriz Godinho Laboratories, 2410-152, Leiria, Portugal
| | - Maria Beatriz Tomaz
- Labeto, Centro de Análises Bioquímicas SA, Beatriz Godinho Laboratories, 2410-152, Leiria, Portugal
| | - André Gonçalves
- Gastroentherology Division, Leiria Central Hospital, Leiria Region Local Health Unit, 2410-197, Leiria, Portugal
| | - Sandra Barbeiro
- Gastroentherology Division, Leiria Central Hospital, Leiria Region Local Health Unit, 2410-197, Leiria, Portugal
| | - Alfonso Benítez-Páez
- Microbiome, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980, Paterna, Valencia, Spain
| | - Sónia Gonçalves Pereira
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541, Leiria, Portugal.
| |
Collapse
|
8
|
Gaber M, Wilson AS, Millen AE, Hovey KM, LaMonte MJ, Wactawski-Wende J, Ochs-Balcom HM, Cook KL. Visceral adiposity in postmenopausal women is associated with a pro-inflammatory gut microbiome and immunogenic metabolic endotoxemia. MICROBIOME 2024; 12:192. [PMID: 39367431 PMCID: PMC11453046 DOI: 10.1186/s40168-024-01901-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 08/06/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Obesity, and in particular abdominal obesity, is associated with an increased risk of developing a variety of chronic diseases. Obesity, aging, and menopause are each associated with differential shifts in the gut microbiome. Obesity causes chronic low-grade inflammation due to increased lipopolysaccharide (LPS) levels which is termed "metabolic endotoxemia." We examined the association of visceral adiposity tissue (VAT) area, circulating endotoxemia markers, and the gut bacterial microbiome in a cohort of aged postmenopausal women. METHODS Fifty postmenopausal women (mean age 78.8 ± 5.3 years) who had existing adipose measurements via dual x-ray absorptiometry (DXA) were selected from the extremes of VAT: n = 25 with low VAT area (45.6 ± 12.5 cm2) and n = 25 with high VAT area (177.5 ± 31.3 cm2). Dietary intake used to estimate the Healthy Eating Index (HEI) score was assessed with a food frequency questionnaire. Plasma LPS, LPS-binding protein (LBP), anti-LPS antibodies, anti-flagellin antibodies, and anti-lipoteichoic acid (LTA) antibodies were measured by ELISA. Metagenomic sequencing was performed on fecal DNA. Female C57BL/6 mice consuming a high-fat or low-fat diet were treated with 0.4 mg/kg diet-derived fecal isolated LPS modeling metabolic endotoxemia, and metabolic outcomes were measured after 6 weeks. RESULTS Women in the high VAT group showed increased Proteobacteria abundance and a lower Firmicutes/Bacteroidetes ratio. Plasma LBP concentration was positively associated with VAT area. Plasma anti-LPS, anti-LTA, and anti-flagellin IgA antibodies were significantly correlated with adiposity measurements. Women with high VAT showed significantly elevated LPS-expressing bacteria compared to low VAT women. Gut bacterial species that showed significant associations with both adiposity and inflammation (anti-LPS IgA and LBP) were Proteobacteria (Escherichia coli, Shigella spp., and Klebsiella spp.) and Veillonella atypica. Healthy eating index (HEI) scores negatively correlated with % body fat and anti-LPS IgA antibodies levels. Preclinical murine model showed that high-fat diet-fed mice administered a low-fat diet fecal-derived LPS displayed reduced body weight, decreased % body fat, and improved glucose tolerance test parameters when compared with saline-injected or high-fat diet fecal-derived LPS-treated groups consuming a high-fat diet. CONCLUSIONS Increased VAT in postmenopausal women is associated with elevated gut Proteobacteria abundance and immunogenic metabolic endotoxemia markers. Low-fat diet-derived fecal-isolated LPS improved metabolic parameters in high-fat diet-fed mice giving mechanistic insights into potential pro-health signaling mediated by under-acylated LPS isoforms. Video Abstract.
Collapse
Grants
- W81XWH-20-1-0014 Congressionally Directed Medical Research Programs
- W81XWH-20-1-0014 Congressionally Directed Medical Research Programs
- W81XWH-20-1-0014 Congressionally Directed Medical Research Programs
- R01 DE013505, R01 DE024523 NIDCR NIH HHS
- R01 DE013505, R01 DE024523 NIDCR NIH HHS
- R01 DE013505, R01 DE024523 NIDCR NIH HHS
- R01 DE013505, R01 DE024523 NIDCR NIH HHS
- R01 DE013505, R01 DE024523 NIDCR NIH HHS
- HHSN268201600018C, HHSN268201600001C, HHSN268201600002C, HHSN268201600003C, and HHSN268201600004C NHLBI NIH HHS
- HHSN268201600018C, HHSN268201600001C, HHSN268201600002C, HHSN268201600003C, and HHSN268201600004C NHLBI NIH HHS
Collapse
Affiliation(s)
- Mohamed Gaber
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Amy E Millen
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Kathleen M Hovey
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Michael J LaMonte
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Heather M Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA.
| | - Katherine L Cook
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
9
|
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
|
10
|
Iorfida D, Valitutti F, Vestri A, D'Adamo G, Passaro T, Crocco M, Malerba F, Monzani A, Rabbone I, Pensabene L, Giancotti L, Graziano F, Citrano M, Ferretti F, Trovato CM, Pacenza C, Iasevoli M, Banzato C, Lubrano R, Montuori M. Prevalence of delivery mode in an Italian nationwide cohort with celiac disease: a SIGENP multicenter retrospective study (the CD-deliver-IT). Ital J Pediatr 2024; 50:129. [PMID: 39061072 PMCID: PMC11282831 DOI: 10.1186/s13052-024-01710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Studies have indicated an association between cesarean section (CS), especially elective CS, and an increased risk of celiac disease (CD), but the conclusions of other studies are contradictory. The primary aim of this study (CD-deliver-IT) was to evaluate the rate of CS in a large population of CD patients throughout Italy. METHODS: This national multicenter retrospective study was conducted between December 2020 and November 2021. The coordinating center was the Pediatric Gastroenterology and Liver Unit of Policlinico Umberto I, Sapienza, University of Rome, Lazio, Italy. Eleven other referral centers for CD have participated to the study. Each center has collected data on mode of delivery and perinatal period of all CD patients referring to the center in the last 40 years. RESULTS Out of 3,259 CD patients recruited in different Italian regions, data on the mode of delivery were obtained from 3,234. One thousand nine hundred forty-one (1,941) patients (60%) were born vaginally and 1,293 (40%) by CS (8.3% emergency CS, 30.1% planned CS, 1.5% undefined CS). A statistically significant difference was found comparing median age at time of CD diagnosis of patients who were born by emergency CS (4 years, CI 95% 3.40-4.59), planned CS (7 years, CI 95% 6.02-7.97) and vaginal delivery (6 years, CI 95% 5.62-6.37) (log rank p < 0.0001). CONCLUSIONS This is the first Italian multicenter study aiming at evaluating the rate of CS in a large population of CD patients through Italy. The CS rate found in our CD patients is higher than rates reported in the general population over the last 40 years and emergency CS seems to be associated with an earlier onset of CD compared to vaginal delivery or elective CS in our large nationwide retrospective cohort. This suggests a potential role of the mode of delivery on the risk of developing CD and on its age of onset, but it is more likely that it works in concert with other perinatal factors. Further prospective studies on other perinatal factors potentially influencing gut microbiota are awaited in order to address heavy conflicting evidence reaming in this research field.
Collapse
Affiliation(s)
- Donatella Iorfida
- Department of Maternal and Child Health, Pediatrics and Neonatology Unit, Santa Maria Goretti Hospital, Sapienza - University of Rome, Latina, Italy
| | - Francesco Valitutti
- Department of Surgical and Biomedical Sciences, Pediatric Clinic, University of Perugia, Perugia, Italy
| | - Annarita Vestri
- Department of Public Health and Infectious Disease, Sapienza - University of Rome, Rome, Italy
| | - Grazia D'Adamo
- Pediatric Unit, AOU Salerno, P.O. Cava de' Tirreni, Salerno, Italy
| | - Tiziana Passaro
- Pediatric Unit, AOU Salerno, P.O. Cava de' Tirreni, Salerno, Italy
| | - Marco Crocco
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Federica Malerba
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Alice Monzani
- Department of Health Sciences, Division of Paediatrics, University of Piemonte Orientale, Novara, Italy
| | - Ivana Rabbone
- Department of Health Sciences, Division of Paediatrics, University of Piemonte Orientale, Novara, Italy
| | - Licia Pensabene
- Department of Surgical and Medical Sciences, Pediatric Unit, Magna Graecia University, Catanzaro, Italy
| | - Laura Giancotti
- Department of Surgical and Medical Sciences, Pediatric Unit, Magna Graecia University, Catanzaro, Italy
| | | | - Michele Citrano
- Pediatric Unit, Villa Sofia - Cervello Hospital, Palermo, Italy
| | - Francesca Ferretti
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy
| | - Chiara Maria Trovato
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy
| | | | - Mario Iasevoli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Pediatrics Section, University of Salerno, Baronissi, Italy
| | | | - Riccardo Lubrano
- Department of Maternal and Child Health, Pediatrics and Neonatology Unit, Santa Maria Goretti Hospital, Sapienza - University of Rome, Latina, Italy
| | - Monica Montuori
- Maternal and Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza - University of Rome, Rome, Italy.
| |
Collapse
|
11
|
Matera M, Guandalini S. How the Microbiota May Affect Celiac Disease and What We Can Do. Nutrients 2024; 16:1882. [PMID: 38931237 PMCID: PMC11206804 DOI: 10.3390/nu16121882] [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: 05/21/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Celiac disease (CeD) is an autoimmune disease with a strong association with human leukocyte antigen (HLA), characterized by the production of specific autoantibodies and immune-mediated enterocyte killing. CeD is a unique autoimmune condition, as it is the only one in which the environmental trigger is known: gluten, a storage protein present in wheat, barley, and rye. How and when the loss of tolerance of the intestinal mucosa to gluten occurs is still unknown. This event, through the activation of adaptive immune responses, enhances epithelial cell death, increases the permeability of the epithelial barrier, and induces secretion of pro-inflammatory cytokines, resulting in the transition from genetic predisposition to the actual onset of the disease. While the role of gastrointestinal infections as a possible trigger has been considered on the basis of a possible mechanism of antigen mimicry, a more likely alternative mechanism appears to involve a complex disruption of the gastrointestinal microbiota ecosystem triggered by infections, rather than the specific effect of a single pathogen on intestinal mucosal homeostasis. Several lines of evidence show the existence of intestinal dysbiosis that precedes the onset of CeD in genetically at-risk subjects, characterized by the loss of protective bacterial elements that both epigenetically and functionally can influence the response of the intestinal epithelium leading to the loss of gluten tolerance. We have conducted a literature review in order to summarize the current knowledge about the complex and in part still unraveled dysbiosis that precedes and accompanies CeD and present some exciting new data on how this dysbiosis might be prevented and/or counteracted. The literature search was conducted on PubMed.gov in the time frame 2010 to March 2024 utilizing the terms "celiac disease and microbiota", "celiac disease and microbiome", and "celiac disease and probiotics" and restricting the search to the following article types: Clinical Trials, Meta-Analysis, Review, and Systematic Review. A total of 364 papers were identified and reviewed. The main conclusions of this review can be outlined as follows: (1) quantitative and qualitative changes in gut microbiota have been clearly documented in CeD patients; (2) intestinal microbiota's extensive and variable interactions with enterocytes, viral and bacterial pathogens and even gluten combine to impact the inflammatory immune response to gluten and the loss of gluten tolerance, ultimately affecting the pathogenesis, progression, and clinical expression of CeD; (3) gluten-free diet fails to restore the eubiosis of the digestive tract in CeD patients, and also negatively affects microbial homeostasis; (4) new tools allowing targeted microbiota therapy, such as the use of probiotics (a good example being precision probiotics like the novel strain of B. vulgatus (20220303-A2) begin to show exciting potential applications.
Collapse
Affiliation(s)
- Mariarosaria Matera
- Pediatric Clinical Microbiomics Service, Misericordia Hospital, Via Senese 161, 58100 Grosseto, Italy;
| | - Stefano Guandalini
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Celiac Disease Center, University of Chicago Medicine, 5841 S. Maryland Ave. MC 4065, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Galipeau HJ, Hinterleitner R, Leonard MM, Caminero A. Non-Host Factors Influencing Onset and Severity of Celiac Disease. Gastroenterology 2024; 167:34-50. [PMID: 38286392 PMCID: PMC11653303 DOI: 10.1053/j.gastro.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts; Center for Celiac Research and Treatment, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
13
|
Lin Y, Xie Z, Li Z, Yuan C, Zhang C, Li Y, Xie K, Wang K. The microbiota-gut-brain axis: A crucial immunomodulatory pathway for Bifidobacterium animalis subsp. lactis' resilience against LPS treatment in neonatal rats. Int J Biol Macromol 2024; 266:131255. [PMID: 38556221 DOI: 10.1016/j.ijbiomac.2024.131255] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
An imbalanced gut microflora may contribute to immune disorders in neonates due to an immature gut barrier. Bacterial toxins, particularly, can trigger the immune system, potentially resulting in uncontrolled gut and systemic inflammation. Previous research has revealed that Bifidobacterium animalis subsp. lactis (B. lactis) could protect against early-life pathogen infections by enhancing the gut barrier. However, the effects of B. lactis on a compromised immune system remain uncertain. Hence, this study concentrated on the immunomodulatory effects and mechanisms of B. lactis in neonatal rats intraperitoneally injected with lipopolysaccharide (LPS), a bacterial toxin and inflammatory mediator. First, B. lactis significantly alleviated the adverse effects induced by LPS on the growth, development, and body temperature of neonatal rats. Second, B. lactis significantly reduced the immune responses and damage induced by LPS, affecting both systemic and local immune responses in the peripheral blood, gut, and brain. Notably, B. lactis exhibited extra potent neuroprotective and neurorepair effects. Our research found that pre-treatment with B. lactis shaped the diverse gut microecology by altering both microbial populations and metabolic biomolecules, closely linked to immunomodulation. Overall, this study elucidated the multifaceted roles of B. lactis in neonatal hosts against pathogenic infection and immune disorder, revealing the existence of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Yugui Lin
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China; Department of Microbiology, Guangxi Medical University, 530021 Nanning, China.
| | - Zhong Xie
- Department of Microbiology, Guangxi Medical University, 530021 Nanning, China
| | - Zhouyi Li
- Department of Microbiology, Guangxi Medical University, 530021 Nanning, China
| | - Chunlei Yuan
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China
| | - Chilun Zhang
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China
| | - Yanfen Li
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China
| | - Kunke Xie
- Immunology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China
| | - Ke Wang
- Immunology Laboratory, Zhongshan Bo'ai Hospital, 528400 Zhongshan, China
| |
Collapse
|
14
|
Tran T, Senger S, Baldassarre M, Brosnan RA, Cristofori F, Crocco M, De Santis S, Elli L, Faherty CS, Francavilla R, Goodchild-Michelman I, Kenyon VA, Leonard MM, Lima RS, Malerba F, Montuori M, Morelli A, Norsa L, Passaro T, Piemontese P, Reed JC, Sansotta N, Valitutti F, Zomorrodi AR, Fasano A. Novel Bacteroides Vulgatus strain protects against gluten-induced break of human celiac gut epithelial homeostasis: a pre-clinical proof-of-concept study. Pediatr Res 2024; 95:1254-1264. [PMID: 38177249 PMCID: PMC11035120 DOI: 10.1038/s41390-023-02960-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Accepted: 11/18/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND AND AIMS We have identified a decreased abundance of microbial species known to have a potential anti-inflammatory, protective effect in subjects that developed Celiac Disease (CeD) compared to those who did not. We aim to confirm the potential protective role of one of these species, namely Bacteroides vulgatus, and to mechanistically establish the effect of bacterial bioproducts on gluten-dependent changes on human gut epithelial functions. METHODS We identified, isolated, cultivated, and sequenced a unique novel strain (20220303-A2) of B. vulgatus found only in control subjects. Using a human gut organoid system developed from pre-celiac patients, we monitored epithelial phenotype and innate immune cytokines at baseline, after exposure to gliadin, or gliadin plus B. vulgatus cell free supernatant (CFS). RESULTS Following gliadin exposure, we observed increases in epithelial cell death, epithelial monolayer permeability, and secretion of pro-inflammatory cytokines. These effects were mitigated upon exposure to B. vulgatus 20220303-A2 CFS, which had matched phenotype gene product mutations. These protective effects were mediated by epigenetic reprogramming of the organoids treated with B. vulgatus CFS. CONCLUSIONS We identified a unique strain of B. vulgatus that may exert a beneficial role by protecting CeD epithelium against a gluten-induced break of epithelial tolerance through miRNA reprogramming. IMPACT Gut dysbiosis precedes the onset of celiac disease in genetically at-risk infants. This dysbiosis is characterized by the loss of protective bacterial strains in those children who will go on to develop celiac disease. The paper reports the mechanism by which one of these protective strains, B. vulgatus, ameliorates the gluten-induced break of gut epithelial homeostasis by epigenetically re-programming the target intestinal epithelium involving pathways controlling permeability, immune response, and cell turnover.
Collapse
Affiliation(s)
- Tina Tran
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stefania Senger
- Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA
| | | | - Rachel A Brosnan
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Fernanda Cristofori
- Pediatric Unit "Bruno Trambusti", Osp Pediatrico Giovanni XXIII, University of Bari, Bari, Italy
| | - Marco Crocco
- Department of Pediatrics, IRCCS Ospedale Giannina Gaslini, Genova, Italy
| | - Stefania De Santis
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Pathology, Case Western University School of Medicine, Cleveland, OH, USA
| | - Luca Elli
- Celiac Disease Referral Center, Ospedale Maggiore Policlinico, Milan, Italy
| | - Christina S Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ruggero Francavilla
- Pediatric Unit "Bruno Trambusti", Osp Pediatrico Giovanni XXIII, University of Bari, Bari, Italy
| | - Isabella Goodchild-Michelman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Victoria A Kenyon
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Maureen M Leonard
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, Mass General for Children, Boston, MA, USA
| | - Rosiane S Lima
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Federica Malerba
- Department of Pediatrics, IRCCS Ospedale Giannina Gaslini, Genova, Italy
| | - Monica Montuori
- Pediatric Gastroenterology Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Annalisa Morelli
- Pediatric Training Program, University of Salerno School of Medicine, Salerno, Italy
| | - Lorenzo Norsa
- Pediatric Hepatology Gastroenterology and Transplant Unit, Ospedale Papa Giovanni XXIII Bergamo, Bergamo, Italy
| | - Tiziana Passaro
- Celiac Disease Referral Center, "San Giovanni di Dio e Ruggi d'Aragona" University Hospital, Pole of Cava de' Tirreni, Salerno, Italy
| | - Pasqua Piemontese
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - James C Reed
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, Mass General for Children, Boston, MA, USA
| | - Naire Sansotta
- Pediatric Hepatology Gastroenterology and Transplant Unit, Ospedale Papa Giovanni XXIII Bergamo, Bergamo, Italy
| | - Francesco Valitutti
- Pediatric Gastroenterology and Liver Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
- European Biomedical Research Institute of Salerno, Salerno, Italy
| | - Ali R Zomorrodi
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, Mass General for Children, Boston, MA, USA.
- European Biomedical Research Institute of Salerno, Salerno, Italy.
| |
Collapse
|
15
|
Zhang L, Agrawal M, Ng SC, Jess T. Early-life exposures and the microbiome: implications for IBD prevention. Gut 2024; 73:541-549. [PMID: 38123972 PMCID: PMC11150004 DOI: 10.1136/gutjnl-2023-330002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
The early-life period is one of microbiome establishment and immune maturation. Early-life exposures are increasingly being recognised to play an important role in IBD risk. The composition of functions of the gut microbiome in the prenatal, perinatal, and postnatal period may be crucial towards development of health or disease, including IBD, later in life. We herein present a comprehensive summary of the interplay between early-life factors and microbiome perturbations, and their association with risk of IBD. In addition, we provide an overview of host and external factors in early life that are known to impact gut microbiome maturation and exposures implicated in IBD risk. Considering the emerging concept of IBD prevention, we propose strategies to minimise maternal and offspring exposure to potentially harmful variables and recommend protective measures during pregnancy and the postpartum period. This holistic view of early-life factors and microbiome signatures among mothers and their offspring will help frame our current understanding of their importance towards IBD pathogenesis and frame the roadmap for preventive strategies.
Collapse
Affiliation(s)
- Lin Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Manasi Agrawal
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York NY, New York, USA
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
16
|
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
|
17
|
Wu K, Bu F, Wu Y, Zhang G, Wang X, He S, Liu MF, Chen R, Yuan H. Exploring noncoding variants in genetic diseases: from detection to functional insights. J Genet Genomics 2024; 51:111-132. [PMID: 38181897 DOI: 10.1016/j.jgg.2024.01.001] [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: 11/05/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
Previous studies on genetic diseases predominantly focused on protein-coding variations, overlooking the vast noncoding regions in the human genome. The development of high-throughput sequencing technologies and functional genomics tools has enabled the systematic identification of functional noncoding variants. These variants can impact gene expression, regulation, and chromatin conformation, thereby contributing to disease pathogenesis. Understanding the mechanisms that underlie the impact of noncoding variants on genetic diseases is indispensable for the development of precisely targeted therapies and the implementation of personalized medicine strategies. The intricacies of noncoding regions introduce a multitude of challenges and research opportunities. In this review, we introduce a spectrum of noncoding variants involved in genetic diseases, along with research strategies and advanced technologies for their precise identification and in-depth understanding of the complexity of the noncoding genome. We will delve into the research challenges and propose potential solutions for unraveling the genetic basis of rare and complex diseases.
Collapse
Affiliation(s)
- Ke Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Fengxiao Bu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Gen Zhang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xin Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Shunmin He
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mo-Fang Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; State Key Laboratory of Molecular Biology, State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Runsheng Chen
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Huijun Yuan
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
18
|
Martínez-Martínez M, Martínez-Martínez M, Soria-Guerra R, Gamiño-Gutiérrez S, Senés-Guerrero C, Santacruz A, Flores-Ramírez R, Salazar-Martínez A, Portales-Pérez D, Bach H, Martínez-Gutiérrez F. Influence of feeding practices in the composition and functionality of infant gut microbiota and its relationship with health. PLoS One 2024; 19:e0294494. [PMID: 38170702 PMCID: PMC10763948 DOI: 10.1371/journal.pone.0294494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024] Open
Abstract
Establishing the infant's gut microbiota has long-term implications on health and immunity. Breastfeeding is recognized as the best practice of infant nutrition in comparison with formula feeding. We evaluated the effects of the primary feeding practices by analyzing the infant growth and the potential association with gut diseases. A cross-sectional and observational study was designed. This study included 55 mothers with infants, who were divided according to their feeding practices in breastfeeding (BF), formula feeding (FF), and combined breast and formula feeding (CF). Anthropometric measurements of the participants were recorded. Additionally, non-invasive fecal samples from the infants were collected to analyze the microbiota by sequencing, immunoglobulin A (IgA) concentration (ELISA), and volatile organic compounds (gas chromatography with an electronic nose). Results showed that the microbiota diversity in the BF group was the highest compared to the other two groups. The IgA levels in the BF group were twice as high as those in the FF group. Moreover, the child´s growth in the BF group showed the best infant development when the data were compared at birth to the recollection time, as noted by the correlation with a decreased concentration of toxic volatile organic compounds. Interestingly, the CF group showed a significant difference in health status when the data were compared with the FF group. We conclude that early health practices influence children's growth, which is relevant to further research about how those infants' health evolved.
Collapse
Affiliation(s)
| | | | - Ruth Soria-Guerra
- Facultad de Ciencias Quimicas, Universidad Autonoma de San Luis Potosí, SLP, Mexico
| | | | | | - Arlette Santacruz
- Posgrado en Biotecnología, Centro de Biotecnología FEMSA, Tecnológico de Monterrey, Monterrey, Mexico
| | - Rogelio Flores-Ramírez
- Laboratorio de Salud Total, Centro de Investigación Aplicada en Ambiente y Salud -CIACYT, Universidad Autónoma de San Luis Potosí, SLP, Mexico
| | | | - Diana Portales-Pérez
- Instituto Mexicano del Seguro Social, Torreón, Mexico
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonoma de San Luis Potosí, SLP, Mexico
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Fidel Martínez-Gutiérrez
- Facultad de Ciencias Quimicas, Universidad Autonoma de San Luis Potosí, SLP, Mexico
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonoma de San Luis Potosí, SLP, Mexico
| |
Collapse
|
19
|
Minot SS, Mayer-Blackwell K, Fiore-Gartland A, Johnson A, Self S, Bhatti P, Yao L, Liu L, Sun X, Jinfa Y, Kublin J. Species- and subspecies-level characterization of health-associated bacterial consortia that colonize the human gut during infancy. Gut Microbes 2024; 16:2414975. [PMID: 39428758 PMCID: PMC11497992 DOI: 10.1080/19490976.2024.2414975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND The human gut microbiome develops rapidly during infancy, a key window of development coinciding with the maturation of the adaptive immune system. However, little is known about the microbiome growth dynamics over the first few months of life and whether there are any generalizable patterns across human populations. We performed metagenomic sequencing on stool samples (n = 94) from a cohort of infants (n = 15) at monthly intervals in the first 6 months of life, augmenting our dataset with seven published studies for a total of 4,441 metagenomes from 1,162 infants. RESULTS Strain-level de novo analysis was used to identify 592 of the most abundant organisms in the infant gut microbiome. Previously unrecognized consortia were identified which exhibited highly correlated abundances across samples and were composed of diverse species spanning multiple genera. Analysis of a published cohort of infants with cystic fibrosis identified one such novel consortium of diverse Enterobacterales which was positively correlated with weight gain. While all studies showed an increased community stability during the first year of life, microbial dynamics varied widely in the first few months of life, both by study and by individual. CONCLUSION By augmenting published metagenomic datasets with data from a newly established cohort, we were able to identify novel groups of organisms that are correlated with measures of robust human development. We hypothesize that the presence of these groups may impact human health in aggregate in ways that individual species may not in isolation.
Collapse
Affiliation(s)
| | | | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Andrew Johnson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Steven Self
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC, Canada
- Epidemiology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, USA
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Lena Yao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Lili Liu
- Key Laboratory of Occupational Disease Prevention and Treatment, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, China
| | - Xin Sun
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yi Jinfa
- Department of Pediatrics, Nanhai Maternity and Child Healthcare Hospital of Foshan, Foshan, China
| | - James Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
- HIV Vaccine Trials Network, Fred Hutchinson Cancer Center, Seattle, USA
| |
Collapse
|
20
|
Fasano A, Matera M. Probiotics to Prevent Celiac Disease and Inflammatory Bowel Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:95-111. [PMID: 39060733 DOI: 10.1007/978-3-031-58572-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The incidence of chronic inflammatory diseases (CIDs) is dramatically increasing in the developed world, resulting in an increased burden of disease in childhood. Currently, there are limited effective strategies for treating or preventing these conditions. To date, myriads of cross-sectional studies have described alterations in the composition of the gut microbiota in a variety of disease states, after the disease has already occurred. We suggest that to mechanically link these microbiome changes with disease pathogenesis, a prospective cohort design is needed to capture changes that precede or coincide with disease onset and symptoms. In addition, these prospective studies must integrate microbiological, metagenomic, meta transcriptomic and metabolomic data with minimal and standardized clinical and environmental metadata that allow to correctly compare and interpret the results of the analysis of the human microbiota in order to build a system-level model of the interactions between the host and the development of the disease. The creation of new biological computational models thus constructed will allow us to finally move from the detection of simple elements of "association" to the identification of elements of real "causality" allowing to provide a mechanistic approach to the exploration of the development of CIDs.This can only be done when these diseases are studied as complex biological networks. In this chapter we discuss the current knowledge regarding the contribution of the microbiome to CID in childhood, focusing on celiac disease and inflammatory bowel disease, with the overall aim of identifying pathways to shift research from descriptive to mechanistic approaches. We then examine how some components of the microbiota, through epigenetic reprogramming, can start the march from genetic predisposition to clinical expression of CIDs, thus opening up new possibilities for intervention, through microbiota therapy targeting the manipulation of the composition and function of the microbiota, for future applications of precision medicine and primary prevention.
Collapse
Affiliation(s)
- Alessio Fasano
- Research Centre for Immunology and Mucosal Biology and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children - Harvard Medical School, Boston, USA, MA.
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition, Mass General for Children - Harvard Medical School, Boston, MA, USA.
| | - Mariarosaria Matera
- Neonatologist, Neurodevelopmental Clinics and Pediatric Clinical Microbiomic - Misericordia Hospital, Grosseto, Italy
| |
Collapse
|
21
|
Zhang A, de Ángel Solá D, Acevedo Flores M, Cao L, Wang L, Kim JG, Tarr PI, Warner BB, Rosario Matos N, Wang L. Infants exposed in utero to Hurricane Maria have gut microbiomes with reduced diversity and altered metabolic capacity. mSphere 2023; 8:e0013423. [PMID: 37754563 PMCID: PMC10597457 DOI: 10.1128/msphere.00134-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/10/2023] [Indexed: 09/28/2023] Open
Abstract
The gut microbiome is a potentially important mechanism that links prenatal disaster exposures with increased disease risks. However, whether prenatal disaster exposures are associated with alterations in the infant's gut microbiome remains unknown. We established a birth cohort study named Hurricane as the Origin of Later Alterations in Microbiome (HOLA) after Hurricane Maria struck Puerto Rico in 2017. We enrolled vaginally born Latino term infants aged 2 to 6 months, including n = 29 infants who were exposed in utero to Hurricane Maria in Puerto Rico and n = 34 infants who were conceived at least 5 months after the hurricane as controls. Shotgun metagenomic sequencing was performed on infant stool swabs. Infants exposed in utero to Hurricane Maria had a reduced diversity in their gut microbiome compared to the control infants, which was mainly seen in the exclusively formula-fed group (P = 0.02). Four bacterial species, including Bacteroides vulgatus, Clostridium innocuum, Bifidobacterium pseudocatenulatum, and Clostridium neonatale, were depleted in the exposure group compared to the control group. Compositional differences in the microbial community and metabolic genes between the exposure and control groups were significant, which were driven by the formula feeding group (P = 0.02 for the microbial community and P = 0.008 for the metabolic genes). Metabolic modules involved in carbohydrate metabolism were reduced in the exposure group. Prenatal maternal exposure to Hurricane Maria was associated with a reduced gut commensal and an altered microbial composition and metabolic potential in the offspring's gut. Breastfeeding can adjust the composition of the gut microbiomes of exposed infants. IMPORTANCE Climate change is a serious issue that is affecting human health. With more frequent and intense weather disasters due to climate change, there is an urgent need to evaluate and understand the impacts of prenatal disaster exposures on the offspring. The prenatal stage is a particularly vulnerable stage for disease origination. However, the impact of prenatal weather disaster exposures on the offspring's gut microbiome has not been evaluated. Our HOLA study starts to fill this knowledge gap and provides novel insights into the microbiome as a mechanism that links prenatal disaster exposures with elevated disease risks. Our major finding that reduced microbial diversity and altered metabolic capacity are associated with prenatal hurricane exposures warrants further studies to evaluate the impact of weather disasters on the unborn.
Collapse
Affiliation(s)
- Ai Zhang
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - David de Ángel Solá
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Midnela Acevedo Flores
- Department of Pediatrics and Obstetrics and Gynecology, San Juan City Hospital Research Unit, San Juan Hospital, San Juan, Puerto Rico
| | - Lijuan Cao
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Leran Wang
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Josh G. Kim
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Phillip I. Tarr
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Barbara B. Warner
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Nicolás Rosario Matos
- Department of Pediatrics and Obstetrics and Gynecology, San Juan City Hospital Research Unit, San Juan Hospital, San Juan, Puerto Rico
| | - Leyao Wang
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Kim K, Kang M, Cho BK. Systems and synthetic biology-driven engineering of live bacterial therapeutics. Front Bioeng Biotechnol 2023; 11:1267378. [PMID: 37929193 PMCID: PMC10620806 DOI: 10.3389/fbioe.2023.1267378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
The past decade has seen growing interest in bacterial engineering for therapeutically relevant applications. While early efforts focused on repurposing genetically tractable model strains, such as Escherichia coli, engineering gut commensals is gaining traction owing to their innate capacity to survive and stably propagate in the intestine for an extended duration. Although limited genetic tractability has been a major roadblock, recent advances in systems and synthetic biology have unlocked our ability to effectively harness native gut commensals for therapeutic and diagnostic purposes, ranging from the rational design of synthetic microbial consortia to the construction of synthetic cells that execute "sense-and-respond" logic operations that allow real-time detection and therapeutic payload delivery in response to specific signals in the intestine. In this review, we outline the current progress and latest updates on microbial therapeutics, with particular emphasis on gut commensal engineering driven by synthetic biology and systems understanding of their molecular phenotypes. Finally, the challenges and prospects of engineering gut commensals for therapeutic applications are discussed.
Collapse
Affiliation(s)
- Kangsan Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Minjeong Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Byung-Kwan Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Graduate School of Engineering Biology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Berryman MA, Ilonen J, Triplett EW, Ludvigsson J. Important denominator between autoimmune comorbidities: a review of class II HLA, autoimmune disease, and the gut. Front Immunol 2023; 14:1270488. [PMID: 37828987 PMCID: PMC10566625 DOI: 10.3389/fimmu.2023.1270488] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Human leukocyte antigen (HLA) genes are associated with more diseases than any other region of the genome. Highly polymorphic HLA genes produce variable haplotypes that are specifically correlated with pathogenically different autoimmunities. Despite differing etiologies, however, many autoimmune disorders share the same risk-associated HLA haplotypes often resulting in comorbidity. This shared risk remains an unanswered question in the field. Yet, several groups have revealed links between gut microbial community composition and autoimmune diseases. Autoimmunity is frequently associated with dysbiosis, resulting in loss of barrier function and permeability of tight junctions, which increases HLA class II expression levels and thus further influences the composition of the gut microbiome. However, autoimmune-risk-associated HLA haplotypes are connected to gut dysbiosis long before autoimmunity even begins. This review evaluates current research on the HLA-microbiome-autoimmunity triplex and proposes that pre-autoimmune bacterial dysbiosis in the gut is an important determinant between autoimmune comorbidities with systemic inflammation as a common denominator.
Collapse
Affiliation(s)
- Meghan A. Berryman
- Triplett Laboratory, Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eric W. Triplett
- Triplett Laboratory, Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Johnny Ludvigsson
- Crown Princess Victoria’s Children’s Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
24
|
Longo M, Jericó D, Córdoba KM, Riezu-Boj JI, Urtasun R, Solares I, Sampedro A, Collantes M, Peñuelas I, Moreno-Aliaga MJ, Ávila MA, Pierro ED, Barajas M, Milagro FI, Dongiovanni P, Fontanellas A. Nutritional Interventions with Bacillus coagulans Improved Glucose Metabolism and Hyperinsulinemia in Mice with Acute Intermittent Porphyria. Int J Mol Sci 2023; 24:11938. [PMID: 37569315 PMCID: PMC10418637 DOI: 10.3390/ijms241511938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Acute intermittent porphyria (AIP) is a metabolic disorder caused by mutations in the porphobilinogen deaminase (PBGD) gene, encoding the third enzyme of the heme synthesis pathway. Although AIP is characterized by low clinical penetrance (~1% of PBGD mutation carriers), patients with clinically stable disease report chronic symptoms and frequently show insulin resistance. This study aimed to evaluate the beneficial impact of nutritional interventions on correct carbohydrate dysfunctions in a mouse model of AIP that reproduces insulin resistance and altered glucose metabolism. The addition of spores of Bacillus coagulans in drinking water for 12 weeks modified the gut microbiome composition in AIP mice, ameliorated glucose tolerance and hyperinsulinemia, and stimulated fat disposal in adipose tissue. Lipid breakdown may be mediated by muscles burning energy and heat dissipation by brown adipose tissue, resulting in a loss of fatty tissue and improved lean/fat tissue ratio. Probiotic supplementation also improved muscle glucose uptake, as measured using Positron Emission Tomography (PET) analysis. In conclusion, these data provide a proof of concept that probiotics, as a dietary intervention in AIP, induce relevant changes in intestinal bacteria composition and improve glucose uptake and muscular energy utilization. Probiotics may offer a safe, efficient, and cost-effective option to manage people with insulin resistance associated with AIP.
Collapse
Affiliation(s)
- Miriam Longo
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (E.D.P.); (P.D.)
| | - Daniel Jericó
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
| | - Karol M. Córdoba
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
| | - José Ignacio Riezu-Boj
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (J.I.R.-B.); (M.J.M.-A.); (F.I.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
| | - Raquel Urtasun
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain; (R.U.); (M.B.)
| | - Isabel Solares
- Rare Disease Unit, Internal Medicine Department, Clinica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Ana Sampedro
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
| | - María Collantes
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- MicroPET Research Unit, CIMA-CUN, 31008 Pamplona, Spain
- Nuclear Medicine-Department, CUN, 31008 Pamplona, Spain
| | - Ivan Peñuelas
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- MicroPET Research Unit, CIMA-CUN, 31008 Pamplona, Spain
- Nuclear Medicine-Department, CUN, 31008 Pamplona, Spain
| | - María Jesús Moreno-Aliaga
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (J.I.R.-B.); (M.J.M.-A.); (F.I.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Matías A. Ávila
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena Di Pierro
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (E.D.P.); (P.D.)
| | - Miguel Barajas
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain; (R.U.); (M.B.)
| | - Fermín I. Milagro
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (J.I.R.-B.); (M.J.M.-A.); (F.I.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (E.D.P.); (P.D.)
| | - Antonio Fontanellas
- Hepatology: Porphyrias & Carcinogenesis Laboratory, Solid Tumors Program, CIMA-University of Navarra, 31008 Pamplona, Spain; (M.L.); (D.J.); (K.M.C.); (A.S.); (M.A.Á.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (M.C.); (I.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
25
|
Jenickova E, Andrén Aronsson C, Mascellani Bergo A, Cinek O, Havlik J, Agardh D. Effects of Lactiplantibacillus plantarum and Lacticaseibacillus paracasei supplementation on the faecal metabolome in children with coeliac disease autoimmunity: a randomised, double-blinded placebo-controlled clinical trial. Front Nutr 2023; 10:1183963. [PMID: 37485388 PMCID: PMC10359497 DOI: 10.3389/fnut.2023.1183963] [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: 03/10/2023] [Accepted: 06/08/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Coeliac disease is a lifelong immune-mediated enteropathy manifested as gluten intolerance in individuals carrying specific human leukocyte antigen (HLA) molecules. Other factors than genetics and gluten intake, however, may play a role in triggering the disease. The gut internal environment is thought to be one of these potential contributing factors, and it can be influenced throughout life. Methods We examine the impact of Lactiplantibacillus plantarum HEAL9 and Lacticaseibacillus paracasei 8700:2 supplementation on the faecal metabolome in genetically predisposed children having tissue transglutaminase autoantibodies, i.e., coeliac disease autoimmunity. Probiotic strains were selected based on their beneficial properties, including mucosal permeability and immune modulation effects. The intervention group (n = 40) and control group (n = 38) took the probiotics or placebo daily for 6 months in a double-blinded randomised trial. Faecal samples were collected at baseline and after 3 and 6 months and analysed using the 1H NMR for metabolome. The incorporation of 16S rRNA sequencing as a supportive dataset complemented the analysis of the metabolome data. Results During the 6 months of intervention, the stool concentrations of 4-hydroxyphenylacetate increased in the intervention group as compared to controls, whereas concentrations of threonine, valine, leucine, isoleucine, methionine, phenylalanine, aspartate, and fumarate decreased. Additionally, a noteworthy effect on the glycine, serine, and threonine metabolic pathway has been observed. Conclusion The findings suggest a modest yet significant impact of the probiotics on the faecal metabolome, primarily influencing proteolytic processes in the gut. Clinical trial registration ClinicalTrials.gov, NCT03176095.
Collapse
Affiliation(s)
- Eliska Jenickova
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | | | - Anna Mascellani Bergo
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | - Ondrej Cinek
- Department of Pediatrics and Department of Medical Microbiology, Charles University in Prague and University Hospital Motol, Prague, Czechia
| | - Jaroslav Havlik
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
26
|
Martín-Masot R, Jiménez-Muñoz M, Herrador-López M, Navas-López VM, Obis E, Jové M, Pamplona R, Nestares T. Metabolomic Profiling in Children with Celiac Disease: Beyond the Gluten-Free Diet. Nutrients 2023; 15:2871. [PMID: 37447198 DOI: 10.3390/nu15132871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Celiac disease (CD) is included in the group of complex or multifactorial diseases, i.e., those caused by the interaction of genetic and environmental factors. Despite a growing understanding of the pathophysiological mechanisms of the disease, diagnosis is still often delayed and there are no effective biomarkers for early diagnosis. The only current treatment, a gluten-free diet (GFD), can alleviate symptoms and restore intestinal villi, but its cellular effects remain poorly understood. To gain a comprehensive understanding of CD's progression, it is crucial to advance knowledge across various scientific disciplines and explore what transpires after disease onset. Metabolomics studies hold particular significance in unravelling the complexities of multifactorial and multisystemic disorders, where environmental factors play a significant role in disease manifestation and progression. By analyzing metabolites, we can gain insights into the reasons behind CD's occurrence, as well as better comprehend the impact of treatment initiation on patients. In this review, we present a collection of articles that showcase the latest breakthroughs in the field of metabolomics in pediatric CD, with the aim of trying to identify CD biomarkers for both early diagnosis and treatment monitoring. These advancements shed light on the potential of metabolomic analysis in enhancing our understanding of the disease and improving diagnostic and therapeutic strategies. More studies need to be designed to cover metabolic profiles in subjects at risk of developing the disease, as well as those analyzing biomarkers for follow-up treatment with a GFD.
Collapse
Affiliation(s)
- Rafael Martín-Masot
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
| | - María Jiménez-Muñoz
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Marta Herrador-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Víctor Manuel Navas-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Elia Obis
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Teresa Nestares
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
- Department of Physiology, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| |
Collapse
|
27
|
Aguayo-Patrón SV, Trujillo-Rivera OA, Cornejo-Granados F, Ochoa-Leyva A, Calderón de la Barca AM. HLA-Haplotypes Influence Microbiota Structure in Northwestern Mexican Schoolchildren Predisposed for Celiac Disease or Type 1 Diabetes. Microorganisms 2023; 11:1412. [PMID: 37374914 DOI: 10.3390/microorganisms11061412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
To contribute to and elucidate the participation of microbiota in celiac disease (CD) and type 1 diabetes (T1D) development, we evaluated the influence of HLA haplotypes, familial risk, and diet on the microbiota of schoolchildren. We conducted a cross-sectional study on 821 apparently healthy schoolchildren, genotyping HLA DQ2/DQ8, and registering familial risk. We analyzed the fecal microbiota using 16S rRNA gene sequencing, and autoantibodies for CD or T1D by ELISA. After analyses, we created three groups: at-high-risk children (Group 1), at-high-risk children plus autoantibodies (Group 2), and nonrisk children (Group 3). HLA influenced the microbiota of Groups 1 and 2, decreasing phylogenetic diversity in comparison to Group 3. The relative abundance of Oscillospiraceae UCG_002, Parabacteroides, Akkermansia, and Alistipes was higher in Group 3 compared to Groups 1 and 2. Moreover, Oscillospiraceae UCG_002 and Parabacteroides were protectors of the autoantibodies' positivity (RRR = 0.441 and RRR = 0.034, respectively). Conversely, Agathobacter was higher in Group 2, and Lachnospiraceae was in both Groups 1 and 2. Lachnospiraceae correlated positively with the sucrose degradation pathway, while the principal genera in Group 3 were associated with amino acid biosynthesis pathways. In summary, HLA and familial risk influence microbiota composition and functionality in children predisposed to CD or T1D, increasing their autoimmunity risk.
Collapse
Affiliation(s)
- Sandra V Aguayo-Patrón
- Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo 83304, Mexico
| | - Omar A Trujillo-Rivera
- Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo 83304, Mexico
| | - Fernanda Cornejo-Granados
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de Mexico, Cuernavaca 62210, Mexico
| | - Adrian Ochoa-Leyva
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de Mexico, Cuernavaca 62210, Mexico
| | - Ana M Calderón de la Barca
- Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo 83304, Mexico
| |
Collapse
|
28
|
Li T, Feng Y, Wang C, Shi T, Abudurexiti A, Zhang M, Gao F. Assessment of causal associations among gut microbiota, metabolites, and celiac disease: a bidirectional Mendelian randomization study. Front Microbiol 2023; 14:1087622. [PMID: 37250054 PMCID: PMC10213403 DOI: 10.3389/fmicb.2023.1087622] [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: 11/02/2022] [Accepted: 04/27/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND A growing number of studies have implicated that gut microbial abundance and metabolite concentration alterations are associated with celiac disease (CD). However, the causal relationship underlying these associations is unclear. Here, we used Mendelian randomization (MR) to reveal the causal effect of gut microbiota and metabolites on CD. METHODS Genome-wide association study (GWAS) summary-level data for gut microbiota, metabolites, and CD were extracted from published GWASs. Causal bacterial taxa and metabolites for CD were determined by two-sample MR analyses. The robustness of the results was assessed with sensitivity analyses. Finally, reverse causality was investigated with a reverse MR analysis. RESULTS Genetically, increased genus Bifidobacterium was potentially associated with higher CD risk (odds ratio [OR] = 1.447, 95% confidence interval [CI]: 1.054-1.988, p = 0.022) while phylum Lentisphaerae (OR = 0.798, 95% CI: 0.648-0.983, p = 0.034) and genus Coprobacter (OR = 0.683, 95% CI: 0.531-0.880, p = 0.003) were related to lower CD risk. Moreover, there were suggestive associations between CD and the following seven metabolites: 1-oleoylglycerophosphoethanolamine, 1-palmitoylglycerophosphoethanolamine, 1,6-anhydroglucose, phenylacetylglutamine, tryptophan betaine, 10-undecenoate, and tyrosine. Sensitivity analyses deemed the results reliable without pleiotropy. CONCLUSION We investigated the causal relationships between gut microbiota, metabolites, and CD with two-sample MR. Our findings suggest several novel potential therapeutic targets for CD treatment. Further understanding of the underlying mechanism may provide insights into CD pathogenesis.
Collapse
Affiliation(s)
- Ting Li
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Yan Feng
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Chun Wang
- Department of Pathology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Tian Shi
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Adilai Abudurexiti
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Mengxia Zhang
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Feng Gao
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, Xinjiang, China
| |
Collapse
|
29
|
Leonard MM, Kenyon V, Valitutti F, Pennacchio-Harrington R, Piemontese P, Francavilla R, Norsa L, Passaro T, Crocco M, Baldassarre M, Trovato CM, Fasano A, the CDGEMM working group. Cohort profile: Celiac disease genomic, environmental, microbiome and metabolome study; a prospective longitudinal birth cohort study of children at-risk for celiac disease. PLoS One 2023; 18:e0282739. [PMID: 36888627 PMCID: PMC9994673 DOI: 10.1371/journal.pone.0282739] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/21/2023] [Indexed: 03/09/2023] Open
Abstract
The Celiac Disease Genomic, Environmental, Microbiome and Metabolomic (CDGEMM) study is an international prospective birth cohort in children at-risk of developing celiac disease (CD). The CDGEMM study has been designed to take a multi-omic approach to predicting CD onset in at-risk individuals. Participants are required to have a first-degree family member with biopsy diagnosed CD and must be enrolled prior to the introduction of solid food. Participation involves providing blood and stool samples longitudinally over a period of five years as well as answering questionnaires related to the participant, their family, and environment. Recruitment and data collection have been ongoing since 2014. As of 2022 we have a total of 554 participants and the average age of the cohort is 56.4 months. A total of 54 participants have developed positive antibodies for CD and 31 have confirmed CD. Approximately 80% of the 54 participants with CD have developed it by 3 years of age. To date we have identified several microbial strains, pathways, and metabolites occurring in increased abundance and detected before CD onset, which have previously been linked to autoimmune and inflammatory conditions while others occurred in decreased abundance before CD onset and are known to have anti-inflammatory effects. Our ongoing analysis includes expanding our metagenomic and metabolomic analyses, evaluating environmental risk factors linked to CD onset, and mechanistic studies investigating how alterations in the microbiome and metabolites may protect against or contribute to CD development.
Collapse
Affiliation(s)
- Maureen M. Leonard
- Mass General Hospital for Children and Division of Pediatric Gastroenterology and Nutrition, Harvard Medical School, Boston, Massachusetts, United States of America
- Celiac Research Program, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Victoria Kenyon
- Celiac Research Program, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Francesco Valitutti
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
- Pediatric Unit, Maternal and Child Health Department, AOU San Giovanni di Dio e Ruggi d’Aragona, Salerno, Italy
| | | | - Pasqua Piemontese
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Ruggiero Francavilla
- Pediatric Unit " Bruno Trambusti", Osp Pediatrico Giovanni XXIII, University of Bari, Bari, Italy
| | - Lorenzo Norsa
- Pediatric Hepatology Gastroenterology and Transplant Unit, Ospedale Papa Giovanni XXIII Bergamo, Bergamo, Italy
| | - Tiziana Passaro
- Celiac Disease Referral Center, “San Giovanni di Dio e Ruggi d’Aragona” University Hospital, Pole of Cava de’ Tirreni, Salerno, Italy
| | - Marco Crocco
- Pediatrics, IRCCS Ospedale Giannina Gaslini, Genova, Italy
| | | | | | - Alessio Fasano
- Mass General Hospital for Children and Division of Pediatric Gastroenterology and Nutrition, Harvard Medical School, Boston, Massachusetts, United States of America
- Celiac Research Program, Harvard Medical School, Boston, Massachusetts, United States of America
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | |
Collapse
|
30
|
Rossi RE, Dispinzieri G, Elvevi A, Massironi S. Interaction between Gut Microbiota and Celiac Disease: From Pathogenesis to Treatment. Cells 2023; 12:823. [PMID: 36980164 PMCID: PMC10047417 DOI: 10.3390/cells12060823] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/24/2022] [Accepted: 01/01/2023] [Indexed: 03/09/2023] Open
Abstract
Celiac disease (CD) is a common systemic disorder that results from an abnormal response of human immunity to gluten intake, affecting the small intestine. In individuals who carry a genetic susceptibility, CD is triggered by environmental factors, including viral infections and dysbiosis of the gut microbiota. The gut microbiome is essential in controlling the immune system, and recent findings indicate that changes in the gut microbiome may contribute to various chronic immune disorders, such as CD through mechanisms that still require further exploration. Some bacteria exhibit epitopes that mimic gliadin and may enhance an immune response in the host. Other bacteria, including Pseudomonas aeruginosa, may work in conjunction with gluten to trigger and escalate intestinal inflammation. The microbiota may also directly influence antigen development through the production of immunogenic or tolerogenic gluten peptides or directly influence intestinal permeability through the release of zonulin. Finally, the gut microbiome can impact intestinal inflammation by generating proinflammatory or anti-inflammatory cytokines and metabolites. It is crucial to consider the impact of genetic factors (specifically, HLA-DQ haplotypes), perinatal elements such as birth mode, type of infant feeding, and antibiotic and infection exposure on the composition of the early intestinal microbiome. According to the available studies, the gut microbiome alterations associated with CD tend to exhibit a decreased presence of beneficial bacteria, including some anti-inflammatory Bifidobacterium species. However, some controversy remains as some reports have found no significant differences between the gut microbiomes of individuals with and without CD. A better understanding of the gut microbiome's role in the development of CD would greatly benefit both prevention and treatment efforts, especially in complicated or treatment-resistant cases. Here, we have attempted to summarize the available evidence on the relationship between the gut microbiota and CD, with a particular focus on potential therapeutic targets.
Collapse
Affiliation(s)
- Roberta Elisa Rossi
- Gastroenterology and Endoscopy Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Giulia Dispinzieri
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca and European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Via Pergolesi 33, 20900 Monza, Italy
| | - Alessandra Elvevi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca and European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Via Pergolesi 33, 20900 Monza, Italy
| | - Sara Massironi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca and European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Via Pergolesi 33, 20900 Monza, Italy
| |
Collapse
|
31
|
Ivanova M, Bottiglieri L, Sajjadi E, Venetis K, Fusco N. Malignancies in Patients with Celiac Disease: Diagnostic Challenges and Molecular Advances. Genes (Basel) 2023; 14:376. [PMID: 36833303 PMCID: PMC9956047 DOI: 10.3390/genes14020376] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Celiac disease (CD) is a multiorgan autoimmune disorder of the chronic intestinal disease group characterized by duodenal inflammation in genetically predisposed individuals, precipitated by gluten ingestion. The pathogenesis of celiac disease is now widely studied, overcoming the limits of the purely autoimmune concept and explaining its hereditability. The genomic profiling of this condition has led to the discovery of numerous genes involved in interleukin signaling and immune-related pathways. The spectrum of disease manifestations is not limited to the gastrointestinal tract, and a significant number of studies have considered the possible association between CD and neoplasms. Patients with CD are found to be at increased risk of developing malignancies, with a particular predisposition of certain types of intestinal cancer, lymphomas, and oropharyngeal cancers. This can be partially explained by common cancer hallmarks present in these patients. The study of gut microbiota, microRNAs, and DNA methylation is evolving to find the any possible missing links between CD and cancer incidence in these patients. However, the literature is extremely mixed and, therefore, our understanding of the biological interplay between CD and cancer remains limited, with significant implications in terms of clinical management and screening protocols. In this review article, we seek to provide a comprehensive overview of the genomics, epigenomics, and transcriptomics data on CD and its relation to the most frequent types of neoplasms that may occur in these patients.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Luca Bottiglieri
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
32
|
Girdhar K, Dogru YD, Huang Q, Yang Y, Tolstikov V, Raisingani A, Chrudinova M, Oh J, Kelley K, Ludvigsson JF, Kiebish MA, Palm NW, Ludvigsson J, Altindis E. Dynamics of the gut microbiome, IgA response, and plasma metabolome in the development of pediatric celiac disease. MICROBIOME 2023; 11:9. [PMID: 36639805 PMCID: PMC9840338 DOI: 10.1186/s40168-022-01429-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Celiac disease (CD) is an autoimmune disorder triggered by gluten consumption. Almost all CD patients possess human leukocyte antigen (HLA) DQ2/DQ8 haplotypes; however, only a small subset of individuals carrying these alleles develop CD, indicating the role of environmental factors in CD pathogenesis. The main objective of this study was to determine the contributory role of gut microbiota and microbial metabolites in CD onset. To this end, we obtained fecal samples from a prospective cohort study (ABIS) at ages 2.5 and 5 years. Samples were collected from children who developed CD after the final sample collection (CD progressors) and healthy children matched by age, HLA genotype, breastfeeding duration, and gluten-exposure time (n=15-16). We first used 16S sequencing and immunoglobulin-A sequencing (IgA-seq) using fecal samples obtained from the same children (i) 16 controls and 15 CD progressors at age 2.5 and (ii) 13 controls and 9 CD progressors at age 5. We completed the cytokine profiling, and plasma metabolomics using plasma samples obtained at age 5 (n=7-9). We also determined the effects of one microbiota-derived metabolite, taurodeoxycholic acid (TDCA), on the small intestines and immune cell composition in vivo. RESULTS CD progressors have a distinct gut microbiota composition, an increased IgA response, and unique IgA targets compared to healthy subjects. Notably, 26 plasma metabolites, five cytokines, and one chemokine were significantly altered in CD progressors at age 5. Among 26 metabolites, we identified a 2-fold increase in TDCA. TDCA treatment alone caused villous atrophy, increased CD4+ T cells, Natural Killer cells, and two important immunoregulatory proteins, Qa-1 and NKG2D expression on T cells while decreasing T-regulatory cells in intraepithelial lymphocytes (IELs) in C57BL/6J mice. CONCLUSIONS Pediatric CD progressors have a distinct gut microbiota composition, plasma metabolome, and cytokine profile before diagnosis. Furthermore, CD progressors have more IgA-coated bacteria and unique targets of IgA in their gut microbiota. TDCA feeding alone stimulates an inflammatory immune response in the small intestines of C57BJ/6 mice and causes villous atrophy, the hallmark of CD. Thus, a microbiota-derived metabolite, TDCA, enriched in CD progressors' plasma, has the potential to drive inflammation in the small intestines and enhance CD pathogenesis. Video Abstract.
Collapse
Affiliation(s)
- Khyati Girdhar
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | | | - Qian Huang
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | | | - Amol Raisingani
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | | | - Jaewon Oh
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Kristina Kelley
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
| | | | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, 58185, Linköping, SE, Sweden
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
33
|
Xu J, Fang H, Chong Y, Lin L, Xie T, Ji J, Shen C, Shi C, Shan J. Cyclophosphamide Induces Lipid and Metabolite Perturbation in Amniotic Fluid during Rat Embryonic Development. Metabolites 2022; 12:1105. [PMID: 36422245 PMCID: PMC9693482 DOI: 10.3390/metabo12111105] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/09/2022] [Indexed: 08/13/2023] Open
Abstract
Cyclophosphamide (CP) has been proven to be an embryo-fetal toxic. However, the mechanism responsible for the toxicity of the teratogenic agent has not been fully explored. This study aimed to examine the teratogenicity of CP when administered in the sensitive period of pregnant rats. The effect of CP on the lipid and metabolic profiles of amniotic fluid was evaluated using a UHPLC-Q-Exactive Orbitrap MS-based method. Metabolome analysis was performed using the MS-DIAL software with LipidBlast and NIST. Initially, we identified 636 and 154 lipid compounds in the positive and negative ion modes and 118 metabolites for differential analysis. Mainly 4 types of oxidized lipids in the amniotic fluid were found to accumulate most significantly after CP treatment, including very-long-chain unsaturated fatty acids (VLCUFAs), polyunsaturated fatty acid (PUFA)-containing triglycerides (TGs), oxidized phosphatidylcholine (PC), and sphingomyelin (SM). Tryptophan and some long-chain saturated fatty acids were lowered pronouncedly after CP treatment. These findings suggest that CP may exert teratogenic toxicity on pregnant rats through maternal and fetal oxidative stress. The UHPLC-Q-Exactive Orbitrap MS-based lipidomics approach is worthy of wider application for evaluating the potential toxicity of other agents (toxicants) during embryonic development.
Collapse
Affiliation(s)
- Jianya Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Huafeng Fang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Chong
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lili Lin
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tong Xie
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cunsi Shen
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chen Shi
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
34
|
Arcila-Galvis JE, Loria-Kohen V, Ramírez de Molina A, Carrillo de Santa Pau E, Marcos-Zambrano LJ. A comprehensive map of microbial biomarkers along the gastrointestinal tract for celiac disease patients. Front Microbiol 2022; 13:956119. [PMID: 36177469 PMCID: PMC9513315 DOI: 10.3389/fmicb.2022.956119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Dysbiosis of the microbiome has been related to Celiac disease (CeD) progress, an autoimmune disease characterized by gluten intolerance developed in genetically susceptible individuals under certain environmental factors. The microbiome contributes to CeD pathophysiology, modulating the immune response by the action of short-chain fatty acids (SCFA), affecting gut barrier integrity allowing the entrance of gluten-derived proteins, and degrading immunogenic peptides of gluten through endoprolyl peptidase enzymes. Despite the evidence suggesting the implication of gut microbiome over CeD pathogenesis, there is no consensus about the specific microbial changes observed in this pathology. Here, we compiled the largest dataset of 16S prokaryotic ribosomal RNA gene high-throughput sequencing for consensus profiling. We present for the first time an integrative analysis of metataxonomic data from patients with CeD, including samples from different body sites (saliva, pharynx, duodenum, and stool). We found the presence of coordinated changes through the gastrointestinal tract (GIT) characterized by an increase in Actinobacteria species in the upper GIT (pharynx and duodenum) and an increase in Proteobacteria in the lower GIT (duodenum and stool), as well as site-specific changes evidencing a dysbiosis in patients with CeD' microbiota. Moreover, we described the effect of adherence to a gluten-free diet (GFD) evidenced by an increase in beneficial bacteria and a decrease in some Betaproteobacteriales but not fully restoring CeD-related dysbiosis. Finally, we built a Random Forest model to classify patients based on the lower GIT composition achieving good performance.
Collapse
Affiliation(s)
- Juliana Estefanía Arcila-Galvis
- Computational Biology Group, Precision Nutrition, and Cancer Research Program, IMDEA Food Institute, Madrid, Spain.,Computational Epigenomics Laboratory, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Viviana Loria-Kohen
- Nutrition and Clinical Trials Unit, GENYAL Platform IMDEA-Food Institute, Madrid, Spain.,Departamento de Nutrición y Ciencia de los Alimentos, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Ramírez de Molina
- Nutrition and Clinical Trials Unit, GENYAL Platform IMDEA-Food Institute, Madrid, Spain
| | | | - Laura Judith Marcos-Zambrano
- Computational Biology Group, Precision Nutrition, and Cancer Research Program, IMDEA Food Institute, Madrid, Spain
| |
Collapse
|
35
|
El Mouzan M, Al-Hussaini A, Serena G, Assiri A, Al Sarkhy A, Al Mofarreh M, Alasmi M, Fasano A. Microbiota profile of new-onset celiac disease in children in Saudi Arabia. Gut Pathog 2022; 14:37. [PMID: 36076257 PMCID: PMC9461150 DOI: 10.1186/s13099-022-00493-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Background Intestinal dysbiosis has been reported to be associated with celiac disease (CeD) in Western populations but little is known in other populations who have different dietary lifestyle and genetic background. The purpose of this study was to determine whether a different microbiota profile is associated with CeD in children in Saudi Arabia. Results Forty children with CeD, 20 healthy controls, and 19 non-CeD controls were enrolled. The median age at diagnosis was 10.3, 11.3 and 10.6 years in children with CeD, fecal, and mucosal control groups, respectively. Significant differences in microbial composition between children with CeD and controls both at fecal and mucosal level were identified. Fecal samples were more diverse and richer in bacteria as compared with mucosal samples. Proteobacteria were more abundant in duodenal mucosal samples and Firmicutes and Bacteroides were more abundant in stools. The abundance of many taxa was significantly different between children with CeD and non-CeD controls. In mucosal samples, Bifidobacterium angulatum (unadjusted p = 0.006) and Roseburia intestinalis (unadjusted p = 0.031) were examples of most significantly increased species in children with CeD and non-CeD controls, respectively. In fecal samples, there were 169 bacterial species with significantly different abundance between children with CeD and non- CeD controls. Conclusions To our knowledge, this is the first report on the microbial profile in a non-Western population of children with new onset CeD. The fact that mucosal and fecal samples were collected from newly diagnosed children with CeD on normal gluten-containing diet suggests strong association between the identified bacteria and CeD. The identification of many unreported bacterial species significantly associated with CeD, indicates the need for further studies from different populations to expand our understanding of the role of bacteria in the pathogenesis of CeD, hopefully leading to the discovery of new adjuvant treatment options.
Collapse
Affiliation(s)
- Mohammad El Mouzan
- Department of Pediatrics (Gastroenterology), King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Abdulrahman Al-Hussaini
- Department of Pediatrics, Children's Specialist Hospital, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.,Faculty of Medicine, AlFaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Gloria Serena
- Massachusetts General Hospital and Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, Mucosal Immunology and Biology Research Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Asaad Assiri
- Department of Pediatrics (Gastroenterology), King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Al Sarkhy
- Department of Pediatrics (Gastroenterology), King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Al Mofarreh
- Al Mofarreh Polyclinic, King Fahad Medical City, Pediatric Intestinal Failure and Parenteral Nutrition Program, Riyadh, Kingdom of Saudi Arabia
| | - Mona Alasmi
- Department of Pediatrics (Gastroenterology), King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Alessio Fasano
- Massachusetts General Hospital and Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, Mucosal Immunology and Biology Research Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,European Biomedical Research Institute of Salerno, Salerno, Italy
| |
Collapse
|
36
|
Lu M, Feng R, Liu Y, Qin Y, Deng H, Xiao Y, Yin C. Identifying celiac disease-related chemicals by transcriptome-wide association study and chemical-gene interaction analyses. Front Genet 2022; 13:990483. [PMID: 36118884 PMCID: PMC9478571 DOI: 10.3389/fgene.2022.990483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
Celiac disease (CeD) is one of the most common intestinal inflammatory diseases, and its incidence and prevalence have increased over time. CeD affects multiple organs and systems in the body, and environmental factors play a key role in its complex pathogenesis. Although gluten exposure is known to be the causative agent, many unknown environmental factors can trigger or exacerbate CeD. In this study, we investigated the influence of genetic and environmental factors on CeD. Data from a CeD genome-wide association study that included 12,041 CeD cases and 12,228 controls were used to conduct a transcriptome-wide association study (TWAS) using FUSION software. Gene expression reference data were obtained for the small intestine, whole blood, peripheral blood, and lymphocytes. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses using the significant genes identified by the TWAS and conducted a protein–protein interaction network analysis based on the STRING database to detect the function of TWAS-identified genes for CeD. We also performed a chemical-related gene set enrichment analysis (CGSEA) using the TWAS-identified genes to test the relationships between chemicals and CeD. The TWAS identified 8,692 genes, including 101 significant genes (padjusted < 0.05). The CGSEA identified 2,559 chemicals, including 178 chemicals that were significantly correlated with CeD. This study performed a TWAS (for genetic factors) and CGSEA (for environmental factors) and identified several CeD-associated genes and chemicals. The findings expand our understanding of the genetic and environmental factors related to immune-mediated diseases.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Ruoyang Feng
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiao Tong University, Xi’an, China
| | - Yuesheng Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yujie Qin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Hongyang Deng
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
- *Correspondence: Yanfeng Xiao, ; Chunyan Yin,
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
- *Correspondence: Yanfeng Xiao, ; Chunyan Yin,
| |
Collapse
|
37
|
Kumari R, Yadav Y, Misra R, Das U, Das Adhikari U, Malakar P, Dubey GP. Emerging frontiers of antibiotics use and their impacts on the human gut microbiome. Microbiol Res 2022; 263:127127. [PMID: 35914416 DOI: 10.1016/j.micres.2022.127127] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/17/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023]
Abstract
Antibiotics, the primary drugs used to cure bacterial diseases, are increasingly becoming ineffective due to the emergence of multiple drug resistance (MDR) leading to recurrence of previously sensitive pathogens. Human gut microbiome (GM), known to play an important role in various physiological processes, consists of pool of diverse microbes. Indiscriminate use of antibiotics during the life span of an individual may lead to development of resistant microbes e.g. Vibrio, Acinetobacter, Escherichia, Klebsiella, Clostridia, etc. in the human GM. Transmission of antibiotic resistant genes (ARGs) between pathogenic and commensal bacteria occurs more frequently in microbiome communities wherein bacteria communicate and exchange cellular constituents both among themselves and with the host. Additionally, co-factors like 'early vs. late' exposure, type of antibiotics and duration of treatment modulate the adverse effects of antibiotics on GM maturation. Furthermore, factors like mode of birth, ethnicity, malnutrition, demography, diet, lifestyle, etc., which influence GM composition, can also indirectly alter the host response to antibiotics. Currently, advanced 'omics' and culturomics approaches are revealing novel avenues to study the interplay between antibiotics and the microbiome and to identify resistant genes in these bacterial communities. Here, we discuss the recent developments that have given insights into the effects of antibiotics on the homeostatic balance of the gut microbiome and thus on human health.
Collapse
Affiliation(s)
- Rekha Kumari
- Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Yasha Yadav
- Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Richa Misra
- Department of Zoology, Sri Venkateswara College, University of Delhi, Delhi 1100021, India
| | - Utpal Das
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Upasana Das Adhikari
- The Ragon Institute of MGH, MIT and Harvard, 400 Technology Square Cambridge, MA 02139, USA
| | - Pushkar Malakar
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gyanendra P Dubey
- Molecular Microbial Pathogenesis Unit, Institut Pasteur, 28 rue du Docteur Roux, 75724 Cedex 15 Paris, France.
| |
Collapse
|
38
|
Chang D, O’Shea D, Therrien A, Silvester JA. Review article: Becoming and being coeliac-special considerations for childhood, adolescence and beyond. Aliment Pharmacol Ther 2022; 56 Suppl 1:S73-S85. [PMID: 35815825 PMCID: PMC9441244 DOI: 10.1111/apt.16851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 12/09/2022]
Abstract
Classically considered a disease of early childhood characterised by malabsorption and failure to thrive, coeliac disease is now recognised to arise in genetically susceptible individuals at any age. Although permissive HLA genotypes are the strongest predictor of coeliac disease, they are not sufficient. Several prospective cohort studies enrolling genetically at-risk infants have investigated the role of potential triggers of coeliac disease autoimmunity, such as timing of gluten introduction, viral infections and dietary patterns. Much less is known about triggers of coeliac disease in adulthood. Better understanding of factors leading to coeliac disease may be helpful in the management of those with potential coeliac disease (elevated serum celiac antibodies without villous atrophy in the small intestine), many of whom initiate a gluten-free diet without demonstration of villous atrophy. There are a range of clinical presentations of celiac disease in childhood and patterns of coeliac serology, including fluctuation and spontaneous reversion on a gluten-containing diet, vary. There is a current debate over best strategies to manage adults and children with potential coeliac disease to avoid over-treatment and under-treatment. Childhood and adolescence carry unique issues pertaining to the diagnosis and management of coeliac disease, and include nutrition and growth, rescreening, repeat biopsy, dietary adherence concerns and transition to adult care. In conclusion, while coeliac disease has similar pathogenesis and general clinical manifestations in paediatric and adult populations, diagnostic and management approaches need to adapt to the developmental stages.
Collapse
Affiliation(s)
- Denis Chang
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA
| | - Delia O’Shea
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA
| | - Amelie Therrien
- 2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA,3. Beth Israel Deaconess Medical Center, Boston, MA
| | - Jocelyn A Silvester
- 1. Boston Children’s Hospital, Boston, MA,2. Harvard Celiac Research Program, Harvard Medical School, Boston, MA,3. Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
39
|
Calabriso N, Scoditti E, Massaro M, Maffia M, Chieppa M, Laddomada B, Carluccio MA. Non-Celiac Gluten Sensitivity and Protective Role of Dietary Polyphenols. Nutrients 2022; 14:2679. [PMID: 35807860 PMCID: PMC9268201 DOI: 10.3390/nu14132679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Pathogenetically characterized by the absence of celiac disease and wheat allergy, non-celiac gluten sensitivity (NCGS) is a clinical entity triggered by the consumption of gluten-containing foods that relieved by a gluten-free diet. Since it is very difficult to maintain a complete gluten-free diet, there is a high interest in discovering alternative strategies aimed at reducing gluten concentration or mitigating its toxic effects. Plant-based dietary models are usually rich in bioactive compounds, such as polyphenols, recognized to prevent, delay, or even reverse chronic diseases, including intestinal disorders. However, research on the role of polyphenols in mitigating the toxicity of gluten-containing foods is currently limited. We address the metabolic fate of dietary polyphenols, both as free and bound macromolecule-linked forms, with particular reference to the gastrointestinal compartment, where the concentration of polyphenols can reach high levels. We analyze the potential targets of polyphenols including the gluten peptide bioavailability, the dysfunction of the intestinal epithelial barrier, intestinal immune response, oxidative stress and inflammation, and dysbiosis. Overall, this review provides an updated overview of the effects of polyphenols as possible dietary strategies to counteract the toxic effects of gluten, potentially resulting in the improved quality of life of patients with gluten-related disorders.
Collapse
Affiliation(s)
- Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Egeria Scoditti
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (M.M.); (M.C.)
| | - Marcello Chieppa
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (M.M.); (M.C.)
| | - Barbara Laddomada
- Institute of Sciences of Food Production (ISPA), National Research Council (CNR), 73100 Lecce, Italy
| | - Maria Annunziata Carluccio
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| |
Collapse
|
40
|
Catassi C, Verdu EF, Bai JC, Lionetti E. Coeliac disease. Lancet 2022; 399:2413-2426. [PMID: 35691302 DOI: 10.1016/s0140-6736(22)00794-2] [Citation(s) in RCA: 214] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 12/14/2022]
Abstract
Coeliac disease is an autoimmune disorder that primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. Prevalence in the general population ranges from 0·5% to 2%, with an average of about 1%. The development of the coeliac enteropathy depends on a complex immune response to gluten proteins, including both adaptive and innate mechanisms. Clinical presentation of coeliac disease is highly variable and includes classical and non-classical gastrointestinal symptoms, extraintestinal manifestations, and subclinical cases. The disease is associated with a risk of complications, such as osteoporosis and intestinal lymphoma. Diagnosis of coeliac disease requires a positive serology (IgA anti-transglutaminase 2 and anti-endomysial antibodies) and villous atrophy on small-intestinal biopsy. Treatment involves a gluten-free diet; however, owing to the high psychosocial burden of such a diet, research into alternative pharmacological treatments is currently very active.
Collapse
Affiliation(s)
- Carlo Catassi
- Department of Specialized Clinical Sciences and Odontostomatology, Polytechnic University of Marche, Ancona, Italy; Celiac Center and Mucosal Immunology and Biology Research, MassGeneral Hospital for Children-Harvard Medical School, Boston, MA, USA.
| | - Elena F Verdu
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Julio Cesar Bai
- Department of Medicine, Dr C Bonorino Udaondo Gastroenterology Hospital, Buenos Aires, Argentina; Research Institutes, Universidad del Salvador, Buenos Aires, Argentina
| | - Elena Lionetti
- Department of Specialized Clinical Sciences and Odontostomatology, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
41
|
Palmieri O, Castellana S, Bevilacqua A, Latiano A, Latiano T, Panza A, Fontana R, Ippolito AM, Biscaglia G, Gentile A, Gioffreda D, Decina I, Tricarico M, Sinigaglia M, Corbo MR, Mazza T, Perri F, Lamacchia C. Adherence to Gluten-Free Diet Restores Alpha Diversity in Celiac People but the Microbiome Composition Is Different to Healthy People. Nutrients 2022; 14:nu14122452. [PMID: 35745182 PMCID: PMC9228530 DOI: 10.3390/nu14122452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 02/06/2023] Open
Abstract
Celiac disease (CD) is an autoimmune disease with the destruction of small intestinal villi, which occurs in genetically predisposed individuals. At the present moment, a gluten-free diet (GFD) is the only way to restore the functionality of gut mucosa. However, there is an open debate on the effects of long-term supplementation through a GFD, because some authors report an unbalance in microbial taxa composition. Methods: For microbiome analysis, fecal specimens were collected from 46 CD individuals in GFD for at least 2 years and 30 specimens from the healthy controls (HC). Data were analyzed using an ensemble of software packages: QIIME2, Coda-lasso, Clr-lasso, Selbal, PICRUSt2, ALDEx2, dissimilarity-overlap analysis, and dysbiosis detection tests. Results: The adherence to GFD restored the alpha biodiversity of the gut microbiota in celiac people but microbial composition at beta diversity resulted as different to HC. The microbial composition of the CD subjects was decreased in a number of taxa, namely Bifidobacterium longum and several belonging to Lachnospiraceae family, whereas Bacteroides genus was found to be more abundant. Predicted metabolic pathways among the CD bacterial communities revealed an important role in tetrapyrrole biosynthesis. Conclusions: CD patients in GFD had a non-dysbiotic microbial composition for the crude alpha diversity metrics. We found significant differences in beta diversity, in certain taxon, and pathways between subjects with inactive CD in GFD and controls. Collectively, our data may suggest the development of new GFD products by modulating the gut microbiota through diet, supplements of vitamins, and the addition of specific prebiotics.
Collapse
Affiliation(s)
- Orazio Palmieri
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
- Correspondence:
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (S.C.); (T.M.)
| | - Antonio Bevilacqua
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (A.B.); (M.S.); (M.R.C.); (C.L.)
| | - Anna Latiano
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Tiziana Latiano
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Anna Panza
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Rosanna Fontana
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Antonio Massimo Ippolito
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Giuseppe Biscaglia
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Annamaria Gentile
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Domenica Gioffreda
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Ivana Decina
- New Gluten World s.r.l., 71121 Foggia, Italy; (I.D.); (M.T.)
| | | | - Milena Sinigaglia
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (A.B.); (M.S.); (M.R.C.); (C.L.)
| | - Maria Rosaria Corbo
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (A.B.); (M.S.); (M.R.C.); (C.L.)
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (S.C.); (T.M.)
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.L.); (T.L.); (A.P.); (R.F.); (A.M.I.); (G.B.); (A.G.); (D.G.); (F.P.)
| | - Carmela Lamacchia
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (A.B.); (M.S.); (M.R.C.); (C.L.)
- New Gluten World s.r.l., 71121 Foggia, Italy; (I.D.); (M.T.)
| |
Collapse
|
42
|
Liu J, Lundemann AKJ, Reibel J, Pedersen AML. Salivary gland involvement and oral health in patients with coeliac disease. Eur J Oral Sci 2022; 130:e12861. [PMID: 35247226 PMCID: PMC9314853 DOI: 10.1111/eos.12861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/15/2022] [Indexed: 01/25/2023]
Abstract
Coeliac disease (CD) is a chronic immune‐mediated enteropathy triggered by ingestion of gluten. The aim of this study was to investigate if the salivary glands as a component of the mucosal immune system are involved in CD, leading to sialadenitis and salivary gland dysfunction and associated oral manifestations. Twenty patients with CD aged 49.2 (SD 15.5 years) and 20 age‐ and gender‐matched healthy controls underwent an interview regarding general and oral health, serological analysis, a clinical oral examination including bitewing radiographs, Candida smear, assessment of salivary mutans streptococci and lactobacilli levels, unstimulated and chewing‐stimulated whole and parotid saliva flow rates, analysis of secretory IgA, and a labial salivary gland biopsy. Xerostomia, mucosal lesions, dry/cracked lips and focal lymphocytic sialadenitis were more prevalent and extensive in patients with CD than in healthy controls. Moreover, the patients had less gingival inflammation and higher whole saliva flow rates than the healthy controls, but did not differ regarding dental health and levels of cariogenic bacteria and Candida. The major salivary gland function appears unaffected, contributing to maintenance of a balanced microbiota and oral health in CD patients. Xerostomia and labial dryness may be related to minor salivary gland inflammation and subsequent impaired mucosal lubrication.
Collapse
Affiliation(s)
- Jason Liu
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Kristine Juncker Lundemann
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Reibel
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Marie Lynge Pedersen
- Section for Oral Medicine/Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Fornasaro S, Esposito A, Florian F, Pallavicini A, De Leo L, Not T, Lagatolla C, Mezzarobba M, Di Silvestre A, Sergo V, Bonifacio A. Spectroscopic investigation of faeces with surface-enhanced Raman scattering: a case study with coeliac patients on gluten-free diet. Anal Bioanal Chem 2022; 414:3517-3527. [PMID: 35258650 PMCID: PMC9018641 DOI: 10.1007/s00216-022-03975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 11/06/2022]
Abstract
Surface-enhanced Raman scattering (SERS) spectra of faecal samples can be obtained by adding AuNP to their methanol extracts according to the reported protocol, and display bands that are due to bilirubin-like species but also to xanthine and hypoxanthine, two metabolic products secreted by gut bacteria. A total of 27 faecal samples from three different groups, i.e. coeliac patients (n = 9), coeliac patients on gluten-free diet (n = 10) and a control group (n = 8), were characterized with both SERS spectroscopy and 16S rRNA sequencing analysis. Significant differences are present between SERS spectra of coeliac patients and those on gluten-free diet, with a marked increase in the relative intensity of both xanthine and hypoxanthine for the latter. Interestingly, these differences do not correlate with bacterial composition as derived from 16S rRNA sequencing.
Collapse
Affiliation(s)
- Stefano Fornasaro
- Raman Spectroscopy Laboratory, Department of Engineering and Architecture, University of Trieste, P.le Europa 1, 34100, Trieste, Italy
| | - Alessandro Esposito
- Raman Spectroscopy Laboratory, Department of Engineering and Architecture, University of Trieste, P.le Europa 1, 34100, Trieste, Italy
| | - Fiorella Florian
- Department of Life Sciences, University of Trieste, Via Edoardo Weiss 2, 34128, Trieste, TS, Italy
| | - Alberto Pallavicini
- Department of Life Sciences, University of Trieste, Via Edoardo Weiss 2, 34128, Trieste, TS, Italy
| | - Luigina De Leo
- Institute for Maternal Child Health-IRCCS "Burlo Garofolo" Trieste, via dell'Istria 65/1, 34100, Trieste, Italy
| | - Tarcisio Not
- Institute for Maternal Child Health-IRCCS "Burlo Garofolo" Trieste, via dell'Istria 65/1, 34100, Trieste, Italy
| | - Cristina Lagatolla
- Department of Life Sciences, University of Trieste, Via Edoardo Weiss 2, 34128, Trieste, TS, Italy
| | - Marica Mezzarobba
- Department of Life Sciences, University of Trieste, Via Edoardo Weiss 2, 34128, Trieste, TS, Italy
| | - Alessia Di Silvestre
- Raman Spectroscopy Laboratory, Department of Engineering and Architecture, University of Trieste, P.le Europa 1, 34100, Trieste, Italy
| | - Valter Sergo
- Raman Spectroscopy Laboratory, Department of Engineering and Architecture, University of Trieste, P.le Europa 1, 34100, Trieste, Italy
| | - Alois Bonifacio
- Raman Spectroscopy Laboratory, Department of Engineering and Architecture, University of Trieste, P.le Europa 1, 34100, Trieste, Italy.
| |
Collapse
|
44
|
The double-edged sword of gut bacteria in celiac disease and implications for therapeutic potential. Mucosal Immunol 2022; 15:235-243. [PMID: 35031683 DOI: 10.1038/s41385-021-00479-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/22/2021] [Accepted: 12/18/2021] [Indexed: 02/04/2023]
Abstract
Celiac disease (CeD) is an immune-mediated disease, triggered by gluten ingestion, in genetically susceptible individuals. The gluten-free diet (GFD) is the only current treatment for CeD, but is difficult to follow, has high non-adherence rates, and does not always lead to symptomatic or mucosal remission. Microbially-mediated mechanisms have been proposed to contribute to disease pathogenesis, and clinical studies support an association, but mechanistic insight has been difficult to obtain. Recent advances using translational approaches have provided clues to the mechanisms through which bacteria could contribute to CeD pathogenesis. In this review we discuss these bacterially mediated mechanisms, which include the modulation of pathogenic or protective pathways. Targeting these pathways through microbial therapeutics could provide adjuvant therapies to the GFD.
Collapse
|
45
|
Singh A, Kaur H, Midha V, Sood A. Microorganisms in the Pathogenesis and Management of Celiac Disease (CeD). ROLE OF MICROORGANISMS IN PATHOGENESIS AND MANAGEMENT OF AUTOIMMUNE DISEASES 2022:287-307. [DOI: 10.1007/978-981-19-4800-8_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
46
|
Zhao F, Li Z, Dong Z, Wang Z, Guo P, Zhang D, Li S. Exploring the Potential of Exosome-Related LncRNA Pairs as Predictors for Immune Microenvironment, Survival Outcome, and Microbiotain Landscape in Esophageal Squamous Cell Carcinoma. Front Immunol 2022; 13:918154. [PMID: 35880180 PMCID: PMC9307964 DOI: 10.3389/fimmu.2022.918154] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/16/2022] [Indexed: 12/16/2022] Open
Abstract
Accumulating studies have demonstrated the indispensable roles of exosomes and long non-coding RNAs (lncRNAs) in cancer progression and the tumor microenvironment (TME). However, the clinical relevance of exosome-related lncRNAs (ER-lncRNAs) in esophageal squamous cell carcinoma (ESCC) remains unclear. Three subtypes were identified by consensus clustering of 3459 valid ER-lncRNA pairs, of which subtype A is preferentially related to favorable prognosis, lower stromal and immune scores, and higher tumor purity scores. Higher immune cell infiltration, higher mRNA levels of immune checkpoints, higher stromal and immune scores, and lower tumor purity were found in subtype C, which presented a poor prognosis. We developed a prognostic risk score model based on 8 ER-lncRNA pairs in the GEO cohort using univariate Cox regression analysis and LASSO Cox regression analysis. Patients were divided into a high risk-score group and low risk-score group by the cut-off values of the 1-year ROC curves in the training set (GEO cohort) and the validation set (TCGA cohort). Receiver operating characteristic (ROC) curves, Decision curve analysis (DCA), clinical correlation analysis, and univariate and multivariate Cox regression all confirmed that the prognostic model has good predictive power and that the risk score can be used as an independent prognostic factor in different cohorts. By further analyzing the TME based on the risk model, higher immune cell infiltration and more active TME were found in the high-risk group, which presented a poor prognosis. Patients with high risk scores also exhibited higher mRNA levels of immune checkpoints and lower IC50 values, indicating that these patients may be more prone to profit from chemotherapy and immunotherapy. The top five most abundant microbial phyla in ESCC was also identified. The best ER-lncRNAs (AC082651.3, AP000487.1, PLA2G4E-AS1, C8orf49 and AL356056.2) were identified based on machine learning algorithms. Subsequently, the expression levels of the above ER-lncRNAs were analyzed by combining the GTEx and TCGA databases. In addition, qRT-PCR analysis based on clinical samples from our hospital showed a high degree of consistency. This study fills the gap of ER-lncRNA model in predicting the prognosis of patients with ESCC and the risk score-based risk stratification could facilitate the determination of therapeutic option to improve prognoses.
Collapse
Affiliation(s)
- Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine of Hebei Medical University, Shijiazhuang, China
| | - Zhirong Li
- Clinical Laboratory Center, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zefang Dong
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine of Hebei Medical University, Shijiazhuang, China
| | - Zengying Wang
- School of Clinical Medicine of Hebei Medical University, Shijiazhuang, China
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pengfei Guo
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine of Hebei Medical University, Shijiazhuang, China
| | - Dengfeng Zhang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine of Hebei Medical University, Shijiazhuang, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Shujun Li, ; orcid.org/0000-0001-5959-3160
| |
Collapse
|
47
|
Mehrotra I, Serena G, Cetinbas M, Kenyon V, Martin VM, Harshman SG, Zomorrodi AR, Sadreyev RI, Fasano A, Leonard MM. Characterization of the blood microbiota in children with Celiac disease. CURRENT RESEARCH IN MICROBIAL SCIENCES 2021; 2:100069. [PMID: 34841359 PMCID: PMC8610358 DOI: 10.1016/j.crmicr.2021.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 11/11/2022] Open
Abstract
Celiac Disease (CD) is an autoimmune disorder triggered by gluten ingestion that can develop in genetically predisposed individuals. Alterations in the gut microbiota have been suggested to contribute to development of autoimmune conditions including CD. Recent work suggests the existence of a blood microbiota. Evidence that alterations in the blood microbiota potentially influence the development of chronic immune based diseases is increasing. However, there is no published literature regarding the blood microbiota in children, including those with CD. This study aimed to characterize the diversity and taxonomic composition of the blood microbiota of children with CD compared to controls. Whole blood samples were collected from children with active CD, CD in remission, and control subjects and 16S rRNA sequencing was utilized to analyze the blood microbiota. We found 16s rRNA present throughout all pediatric blood samples, providing evidence for the presence of a pediatric blood microbiota. We found significant differences in beta diversity and in abundance of certain taxa (Campylobacterales order, Odoribacteraceae and Helicobacteraceae families, Odoribacter genus and species, and Bacteroides acidifaciens species) between subjects with active CD and controls. These taxa have been previously reported to be associated with immune response and gut-inflammatory diseases. We did not find significant differences between subjects with active and remission CD or between remission CD and controls. Conclusions: We provide evidence for a pediatric blood microbiota and identified higher beta diversity and alterations in the composition of blood microbiota in subjects with active CD compared to controls.
Collapse
Affiliation(s)
- Isha Mehrotra
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
| | - Gloria Serena
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Jackson, 55 Fruit Street, Boston, MA 02114, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
| | - Murat Cetinbas
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Department of Molecular Biology, Massachusetts General Hospital, Simches Research Center, 185 Cambridge Street, CPZN7250 Boston, MA 02114, USA
| | - Victoria Kenyon
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Jackson, 55 Fruit Street, Boston, MA 02114, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
| | - Victoria M. Martin
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Yawkey 6B-6800 Suite, 55 Fruit Street Boston, MA 02114, USA
- Food Allergy Center, Massachusetts General Hospital, MGH Professional Office Building, Suite 530, 275 Cambridge Street Boston, MA 02114, USA
| | - Stephanie G. Harshman
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital: West End Apartments 2 Longfellow Place, Suite 200 Boston, MA 02114, USA
- Neuroendocrine Unit, Massachusetts General Hospital, 100 Blossom St, Boston, MA 02114, USA
| | - Ali R. Zomorrodi
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Jackson, 55 Fruit Street, Boston, MA 02114, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Ruslan I. Sadreyev
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Department of Molecular Biology, Massachusetts General Hospital, Simches Research Center, 185 Cambridge Street, CPZN7250 Boston, MA 02114, USA
| | - Alessio Fasano
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Jackson, 55 Fruit Street, Boston, MA 02114, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Yawkey 6B-6800 Suite, 55 Fruit Street Boston, MA 02114, USA
| | - Maureen M. Leonard
- Center for Celiac Research and Treatment, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Suite 6B, 32 Fruit Street, Boston, MA 02114, USA
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Jackson, 55 Fruit Street, Boston, MA 02114, USA
- Celiac Research Program, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Yawkey Center for Outpatient Care, Yawkey 6B-6800 Suite, 55 Fruit Street Boston, MA 02114, USA
| |
Collapse
|
48
|
Khalkhal E, Rezaei-Tavirani M, Fathi F, Nobakht M. Gh BF, Taherkhani A, Rostami-Nejad M, Asri N, Haidari MH. Screening of Altered Metabolites and Metabolic Pathways in Celiac Disease Using NMR Spectroscopy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1798783. [PMID: 34820452 PMCID: PMC8608527 DOI: 10.1155/2021/1798783] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Celiac disease (CeD) is an autoimmune intestinal disorder caused by gluten protein consumption in genetically predisposed individuals. As biopsy sampling is an invasive procedure, finding novel noninvasive serological markers for screening of at-risk CeD population is a priority. Metabolomics is helpful in monitoring metabolite changes in body fluids and tissues. In the present study, we evaluated serum metabolite levels of CeD patients relative to healthy controls with the aim of introducing new biomarkers for population screening. METHOD We compared the serum metabolic profile of CeD patients (n = 42) and healthy controls (n = 22) using NMR spectroscopy and multivariate analysis. RESULT 25 metabolites were identified by serum metabolic profiling. Levels of 3-hydroxyisobutyric acid and isobutyrate showed significant differences in CeD patients' samples compared with healthy controls (p < 0.05). According to pathway analysis, our data demonstrated that changes in nine metabolic pathways were significantly disrupted/affected in patients with CeD. These enriched pathways are involved in aminoacyl-tRNA biosynthesis; primary bile acid biosynthesis; nitrogen metabolism; glutamine and glutamate metabolism; valine, leucine, and isoleucine biosynthesis and degradation; taurine and hypotaurine metabolism; glyoxylate and dicarboxylate metabolism; glycine, serine, and threonine metabolism; and arginine biosynthesis. CONCLUSION In summary, our results demonstrated that changes in the serum level of 25 metabolites may be useful in distinguishing CeD patients from healthy controls, which have the potential to be considered candidate biomarkers of CeD.
Collapse
Affiliation(s)
- Ensieh Khalkhal
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Fathi
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - B. Fatemeh Nobakht M. Gh
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases 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
| | - Mohammad Hossain Haidari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Olivares M, Flor-Duro A, Sanz Y. Manipulation of the gut microbiome in gluten-intolerance. Curr Opin Clin Nutr Metab Care 2021; 24:536-542. [PMID: 34622826 DOI: 10.1097/mco.0000000000000791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Gluten is a complex mixture of highly immunogenic glutamine- and proline-rich proteins found in some cereals. In celiac disease (CeD), gluten triggers an autoimmune response due to its interaction with the human leukocyte antigen heterodimers that confer the genetic risk. The involvement of gluten in other disorders has also been investigated, but its role beyond CeD is still unclear. Here, we review the most recent evidence of the involvement of gluten in diseases and the opportunities of manipulating the gut microbiota to treat or prevent gluten-related conditions. RECENT FINDINGS Most of the new studies have been conducted in the context of CeD, where important evidence has been gained on associations between the gut microbiota, genotype, and environmental factors such as breastfeeding and antibiotics. The role of the microbiota has been investigated in several prospective, observational and interventional studies with probiotics, which together showed that the gut microbiota could be targeted to ameliorate and aid in the prevention of CeD development. SUMMARY Several studies have evidenced how genetic and environmental factors influence the gut microbiome with consequences in CeD. These findings could inspire the development of microbiota modulation strategies to support the prevention or treatment of CeD.
Collapse
Affiliation(s)
- Marta Olivares
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | | | | |
Collapse
|
50
|
Verdu EF, Schuppan D. Co-factors, Microbes, and Immunogenetics in Celiac Disease to Guide Novel Approaches for Diagnosis and Treatment. Gastroenterology 2021; 161:1395-1411.e4. [PMID: 34416277 DOI: 10.1053/j.gastro.2021.08.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022]
Abstract
Celiac disease (CeD) is a frequent immune-mediated disease that affects not only the small intestine but also many extraintestinal sites. The role of gluten proteins as dietary triggers, HLA-DQ2 or -DQ8 as major necessary genetic predisposition, and tissue transglutaminase (TG2) as mechanistically involved autoantigen, are unique features of CeD. Recent research implicates many cofactors working in synergism with these key triggers, including the intestinal microbiota and their metabolites, nongluten dietary triggers, intestinal barrier defects, novel immune cell phenotypes, and mediators and cytokines. In addition, apart from HLA-DQ2 and -DQ8, multiple and complex predisposing genetic factors and interactions have been defined, most of which overlap with predispositions in other, usually autoimmune, diseases that are linked to CeD. The resultant better understanding of CeD pathogenesis, and its manifold manifestations has already paved the way for novel therapeutic approaches beyond the lifelong strict gluten-free diet, which poses a burden to patients and often does not lead to complete mucosal healing. Thus, supported by improved mouse models for CeD and in vitro organoid cultures, several targeted therapies are in phase 2-3 clinical studies, such as highly effective gluten-degrading oral enzymes, inhibition of TG2, cytokine therapies, induction of tolerance to gluten ingestion, along with adjunctive and preventive approaches using beneficial probiotics and micronutrients. These developments are supported by novel noninvasive markers of CeD severity and activity that may be used as companion diagnostics, allow easy-to perform and reliable monitoring of patients, and finally support personalized therapy for CeD.
Collapse
Affiliation(s)
- Elena F Verdu
- Division of Gastroenterology, Department of Internal Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Detlef Schuppan
- Institute of Translational Immunology,Research Center for Immune Therapy and Celiac Center, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|