Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38719750

ABSTRACT

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Subject(s)
Candida albicans , Celiac Disease , Homeostasis , Mast Cells , Celiac Disease/immunology , Celiac Disease/microbiology , Celiac Disease/metabolism , Humans , Candida albicans/pathogenicity , Candida albicans/immunology , Mast Cells/immunology , Mast Cells/metabolism , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Candidiasis/immunology , Candidiasis/microbiology , Animals , Candida/pathogenicity , Candida/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism
2.
Benef Microbes ; 15(2): 165-177, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653476

ABSTRACT

The changing of microbiome could precede the development of coeliac disease (CeD). We compared the bacterial profile of microbiota of tissues collected simultaneously from the stomach and duodenum in newly diagnosed patients with CeD. Biopsies were collected from 60 children and adolescents aged 2-18 years: (1) 40 patients with CeD; (2) 20 children as control group. The evaluation of the bacterial microbiota was carried out by sequencing the V3-V4 regions of the 16S rRNA subunit, using next-generation sequencing (NGS). The composition of bacterial microbiota was correlated with clinical and blood parameters. The beta diversity analysis revealed a significant dissimilarity in the gastric samples between the CeD and control group (Bray-Curtis index, P = 0.008, and weighted UniFrac distance, P = 0.024). At L2 (phylum level), Campylobacterota was only present in the stomach of the CeD group. A comparison of the abundance of bacteria between the stomach and duodenum showed significant differences in 10 OTUs (operational taxonomic units) in the control and 9 OTUs in the CeD group at L6 (genus) and in 8 OTUs and in 6 OTUs, respectively, at L7 (species). A significant correlation was observed between the genus Novosphingobium in stomach of CeD group and possession of the DQ2.5 and DQ 8 allele, and in the duodenum - between the DQ 8 allele and the species Blautia wexlerae. Significant differences in selected, little-known genera of bacteria suggest their potential role as new biomarkers in the development of CeD. To fully understand the mechanism of CeD development in genetically predisposed individuals, it is necessary to take into account not only the abundance of a given genus or species of bacteria, but also the anatomical location of its occurrence.


Subject(s)
Bacteria , Biomarkers , Celiac Disease , Duodenum , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Stomach , Humans , Celiac Disease/microbiology , Celiac Disease/diagnosis , Child , Pilot Projects , Child, Preschool , Duodenum/microbiology , Duodenum/pathology , Adolescent , Male , RNA, Ribosomal, 16S/genetics , Female , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Stomach/microbiology , Stomach/pathology , High-Throughput Nucleotide Sequencing , Biopsy , DNA, Bacterial/genetics
3.
Virulence ; 15(1): 2334085, 2024 12.
Article in English | MEDLINE | ID: mdl-38528835

ABSTRACT

Candida albicans is an immunogen for anti-Saccharomyces cerevisiae antibodies (ASCA), a serological marker of Crohn's disease. ASCA has also been reported in other autoimmune diseases, including coeliac disease (CeD). A strong antibody response against Hwp1, a protein associated with invasive hyphal form of C. albicans which presents peptide sequence homologies with gliadin, has also been described in CeD. This observation supports the hypothesis that C. albicans hyphal transition in C. albicans may trigger CeD onset through a mechanism of molecular/antigenic mimicry. In this study, we assessed whether the anti-C. albicans oligomannose and anti-Hwp1 protein responses may be linked despite their different pathophysiological significance. The measurement of ASCA levels in a cohort of patients involved in our previous Hwp1 study showed a significant correlation between the two biomarkers. This new observation further reinforces the link between C. albicans and CeD.


Subject(s)
Celiac Disease , Crohn Disease , Humans , Candida albicans/physiology , Celiac Disease/microbiology , Antibodies, Fungal , Antibody Formation
4.
Gut Microbes ; 15(1): 2172955, 2023.
Article in English | MEDLINE | ID: mdl-36751856

ABSTRACT

Current treatment for celiac disease (CD) is adhering to a gluten-free diet (GFD), although its long-term molecular effects are still undescribed. New molecular features detectable in stool may improve and facilitate noninvasive clinical management of CD. For this purpose, fecal small non-coding RNAs (sncRNAs) and gut microbiome profiles were concomitantly explored in CD subjects in relation to strict (or not) GFD adherence over time. In this observational study, we performed small RNA and shotgun metagenomic sequencing in stool from 63 treated CD (tCD) and 3 untreated subjects as well as 66 sex- and age-matched healthy controls. tCD included 51 individuals on strict GFD and with negative transglutaminase (TG) serology (tCD-TG-) and 12 symptomatic with not strict/short-time of GFD adherence and positive TG serology (tCD-TG+). Samples from additional 40 healthy adult individuals and a cohort of 19 untreated pediatric CD subjects and 19 sex/age matched controls were analyzed to further test the outcomes. Several miRNA and microbial profiles were altered in tCD subjects (adj. p < .05). Findings were validated in the external group of adult controls. In tCD-TG-, GFD duration correlated with five miRNA levels (p < .05): for miR-4533-3p and miR-2681-3p, the longer the diet adherence, the less the expression differed from controls. tCD-TG+ and untreated pediatric CD patients showed a similar miRNA dysregulation. Immune-response, trans-membrane transport and cell death pathways were enriched in targets of identified miRNAs. Bifidobacterium longum, Ruminococcus bicirculans, and Haemophilus parainfluenzae abundances shifted (adj. p < .05) with a progressive reduction of denitrification pathways with GFD length. Integrative analysis highlighted 121 miRNA-bacterial relationships (adj. p < .05). Specific molecular patterns in stool characterize CD subjects, reflecting either the long-term GFD effects or the gut inflammatory status, in case of a not strict/short-time adherence. Our findings suggest novel host-microbial interplays and could help the discovery of biomarkers for GFD monitoring over time.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , MicroRNAs , Adult , Humans , Child , Celiac Disease/microbiology , Diet, Gluten-Free , Glutens/adverse effects
5.
Clin Exp Med ; 23(6): 2773-2788, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36609792

ABSTRACT

Gluten-free diet (GFD) is the most effective method to manage celiac disease (CD). Many patients do not reach the complete symptom alleviation, even by strict GFD. Recent studies have reported inconsistent results regarding the beneficial benefits of taking probiotics. Therefore, we aimed to evaluate the effects of probiotics on gastrointestinal (GI) symptoms and the possible underlying causes in CD and celiac disease autoimmunity (CDA) patients. Databases, including PubMed, Scopus, Embase, Web of Science and Google Scholar, were searched for clinical trials published until July 2022 about assessing the effects of probiotics or synbiotics on CD or CDA patients. We collected data on GI symptoms, CD markers, inflammatory and immune responses, adverse events, and gut microbiota. A random effect meta-analysis was used to estimate the pooled standardized mean difference (SMD) and confidence interval (CI). We screened 7234 articles, of which 14 were included in the qualitative analysis and 5 in the quantitative analysis. Probiotics might alleviate GI symptoms, especially in the highly symptomatic patients, and improve immune response in CD and CDA patients. Results of the meta-analysis showed that probiotics increased the abundance of Bifidobacterium (SMD: 0.72, 95%CI (0.13, 1.30) and Lactobacillus (SMD: 0.49, 95%CI (0.18, 0.80) as compared with placebo. Probiotics did not increase the adverse events compared to the placebo. Probiotics might alleviate GI symptoms and immune response and improve dysbiosis in CD and CDA patients. However, high-quality clinical trials are needed to increase the level of evidence. Also, the most suitable combination of probiotics is yet to find.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Probiotics , Humans , Celiac Disease/therapy , Celiac Disease/microbiology , Probiotics/therapeutic use , Diet, Gluten-Free , Dysbiosis/therapy
6.
J Gastroenterol Hepatol ; 37(10): 1844-1852, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35734803

ABSTRACT

BACKGROUND AND AIM: Symptoms of small intestinal bacterial overgrowth (SIBO) and celiac disease (CeD) often overlap, and studies suggest a link between SIBO and CeD. We thus conducted a systematic review and meta-analysis to compare SIBO prevalence in CeD patients and controls and assessed effects of antimicrobial therapy on gastrointestinal symptoms in SIBO positive CeD patients. METHODS: Electronic databases were searched until February 2022 for studies reporting SIBO prevalence in CeD. Prevalence rates, odds ratio (OR), and 95% confidence intervals (CI) of SIBO in CeD and controls were calculated. RESULTS: We included 14 studies, with 742 CeD patients and 178 controls. The pooled prevalence of SIBO in CeD was 18.3% (95% CI: 11.4-28.1), with substantial heterogeneity. Including case-control studies with healthy controls, SIBO prevalence in CeD patients was significantly increased (OR 5.1, 95% CI: 2.1-12.4, P = 0.0001), with minimal heterogeneity. Utilizing breath tests, SIBO prevalence in CeD patients was 20.8% (95% CI: 11.9-33.7), almost two-fold higher compared with culture-based methods at 12.6% (95% CI: 5.1-28.0), with substantial heterogeneity in both analyses. SIBO prevalence in CeD patients nonresponsive to a gluten free diet (GFD) was not statistically higher as compared with those responsive to GFD (OR 1.5, 95% CI: 0.4-5.0, P = 0.511). Antibiotic therapy of SIBO positive CeD patients resulted in improvement in gastrointestinal symptoms in 95.6% (95% CI: 78.0-99.9) and normalization of breath tests. CONCLUSIONS: This study suggests a link between SIBO and CeD. While SIBO could explain nonresponse to a GFD in CeD, SIBO prevalence is not statistically higher in CeD patients non-responsive to GFD. The overall quality of the evidence is low, mainly due to substantial "clinical heterogeneity" and the limited sensitivity/specificity of the available diagnostic tests.


Subject(s)
Celiac Disease , Anti-Bacterial Agents/therapeutic use , Breath Tests , Case-Control Studies , Celiac Disease/complications , Celiac Disease/epidemiology , Celiac Disease/microbiology , Humans , Intestine, Small/microbiology , Prevalence
7.
Sci Rep ; 12(1): 6607, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35459889

ABSTRACT

The potential impact of the composition of maternal breast milk is poorly known in children who develop celiac disease (CD). The aim of our study was to compare the microbiota composition and the concentrations of immune markers in breast milk from mothers whose offspring carried the genetic predisposition to CD, and whether they did or did not develop CD during follow-up for the first 3 years of life. Maternal breast milk samples [CD children (n = 6) and healthy children (n = 18)] were collected 3 months after delivery. Enzyme-linked immunosorbent assays were used to measure TGF-ß1, TGF-ß2, sIgA, MFG-E8 and sCD14. For microbiota analysis, next generation (Illumina) sequencing, real-time PCR and denaturing gradient gel electrophoresis were used. Phylotype abundance and the Shannon 'H' diversity index were significantly higher in breast milk samples in the CD group. There was higher prevalence of the phyla Bacteroidetes and Fusobacteria, the classes Clostridia and Fusobacteriia, and the genera Leptotrichia, Anaerococcus, Sphingomonas, Actynomyces and Akkermansia in the CD group. The immunological markers were differently associated with some Gram-negative bacterial genera and species (Chryseobacterium, Sphingobium) as well as Gram-positive species (Lactobacillus reuteri, Bifidobacterium animalis). In conclusion, the microbiota in breast milk from mothers of genetically predisposed offspring who presented CD showed a higher bacterial phylotype abundance and diversity, as well as a different bacterial composition, as compared with the mothers of unaffected offspring. These immune markers showed some associations with bacterial composition and may influence the risk for development of CD beyond early childhood.


Subject(s)
Celiac Disease , Limosilactobacillus reuteri , Microbiota , Bacteria/genetics , Celiac Disease/microbiology , Child , Child, Preschool , Female , Humans , Lipopolysaccharide Receptors , Milk, Human/microbiology
8.
Am J Surg Pathol ; 46(1): e43-e54, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33739793

ABSTRACT

Celiac disease is a chronic, immune-mediated enteropathy driven by dietary gluten found in genetically susceptible hosts. It has a worldwide distribution, is one of the most common autoimmune disorders globally, and is the only autoimmune condition for which the trigger is known. Despite advances in characterizing mechanisms of disease, gaps in understanding of celiac disease pathogenesis remain. A "frontier" concept is considering what moves an HLA-DQ2 or DQ8-positive individual from asymptomatic gluten tolerance to celiac disease manifestation. In this arena, environmental triggers, including age at the time of initial gluten exposure, the occurrence of usual childhood viral infections, and microbiome alterations have emerged as key events in triggering the symptomatic disease. Pathologists play a major role in frontier aspects of celiac disease. This includes the discovery that duodenal mucosal histology in follow-up biopsies does not correlate with ongoing patient symptoms, antitissue transglutaminase antibody titers and diet adherence in celiac disease patients. Further, in light of recent evidence that the detection of monoclonal T-cell populations in formalin-fixed biopsies is not specific for type II refractory celiac disease, pathologists should resist performing such analyses until common causes of "apparent" refractoriness are excluded. The promise of therapies in celiac disease has led to clinical trials targeting many steps in the inflammatory cascade, which depend upon a pathologist's confirmation of the initial diagnosis and evaluation of responses to therapies. As pathologists continue to be active participants in celiac disease research, partnering with other stakeholders, we will continue to impact this important autoimmune disease.


Subject(s)
Autoimmunity , Celiac Disease/immunology , Duodenum/immunology , Environment , T-Lymphocyte Subsets/immunology , Animals , Bacteria/immunology , Bacteria/pathogenicity , Biopsy , Celiac Disease/microbiology , Celiac Disease/pathology , Celiac Disease/therapy , Duodenum/microbiology , Duodenum/pathology , Dysbiosis , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Phenotype , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors
9.
Dig Dis Sci ; 67(1): 216-223, 2022 01.
Article in English | MEDLINE | ID: mdl-33723701

ABSTRACT

BACKGROUND: Although intestinal fungi are known to interact with the immune system, the relationship between intestinal fungi and childhood celiac disease (CeD), an immune-mediated condition, has rarely been reported. AIMS: The aim of this study was to describe gut fungal profiles in a cohort of children with new-onset CeD. METHODS: Mucosal and fecal samples were collected from children with CeD and controls and subjected to metagenomics analysis of fungal microbiota communities. DNA libraries were sequenced using Illumina HiSeq platform 2 × 150 bp. Bioinformatic analysis was performed to quantify the relative abundance of fungi. Shannon alpha diversity metrics and beta diversity principal coordinate (PCo) analyses were calculated, and DESeq tests were performed between celiac and non-celiac groups. RESULTS: Overall more abundant taxa in samples of children with CeD included Tricholomataceae, Saccharomycetaceae, Saccharomycetes Saccharomyces cerevisiae, and Candida, whereas less abundant taxa included Pichiaceae, Pichia kudriavzevii, Pneumocystis, and Pneumocystis jirovecii. Alpha diversity between CeD and control individuals did not differ significantly, and beta diversity PCo analysis showed overlap of samples from CeD and controls for both fecal or mucosal samples; however, there was a clear separation between mucosal and fecal overall samples CONCLUSIONS: We report fungal dysbiosis in children with CeD, suggesting a possible role in the pathogenesis of CeD. Further larger, controlled, prospective and longitudinal studies are needed to verify the results of this study and clarify the functional role of fungi in CeD.


Subject(s)
Celiac Disease , Dysbiosis , Fungi , Mycobiome , Celiac Disease/diagnosis , Celiac Disease/epidemiology , Celiac Disease/microbiology , Celiac Disease/physiopathology , Child , Dysbiosis/diagnosis , Dysbiosis/microbiology , Feces/microbiology , Female , Fungi/classification , Fungi/immunology , Fungi/isolation & purification , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Metagenomics/methods , Microbiological Phenomena , Mycobiome/genetics , Mycobiome/immunology , Saudi Arabia/epidemiology
10.
Gastroenterology ; 161(5): 1395-1411.e4, 2021 11.
Article in English | MEDLINE | ID: mdl-34416277

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.


Subject(s)
Bacteria/immunology , Celiac Disease/therapy , Gastrointestinal Microbiome , Glutens/immunology , Immunogenetic Phenomena , Immunologic Tests , Intestines/immunology , Animals , Bacteria/pathogenicity , Celiac Disease/genetics , Celiac Disease/immunology , Celiac Disease/microbiology , Disease Models, Animal , Genetic Predisposition to Disease , Host-Pathogen Interactions , Humans , Intestines/microbiology , Intestines/pathology , Phenotype , Predictive Value of Tests , Prognosis , Risk Factors
11.
Nutrients ; 13(7)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34210038

ABSTRACT

The intestinal microbiome may trigger celiac disease (CD) in individuals with a genetic disposition when exposed to dietary gluten. Research demonstrates that nutrition during infancy is crucial to the intestinal microbiome engraftment. Very few studies to date have focused on the breast milk composition of subjects with a history of CD on a gluten-free diet. Here, we utilize a multi-omics approach with shotgun metagenomics to analyze the breast milk microbiome integrated with metabolome profiling of 36 subjects, 20 with CD on a gluten-free diet and 16 healthy controls. These analyses identified significant differences in bacterial and viral species/strains and functional pathways but no difference in metabolite abundance. Specifically, three bacterial strains with increased abundance were identified in subjects with CD on a gluten-free diet of which one (Rothia mucilaginosa) has been previously linked to autoimmune conditions. We also identified five pathways with increased abundance in subjects with CD on a gluten-free diet. We additionally found four bacterial and two viral species/strains with increased abundance in healthy controls. Overall, the differences observed in bacterial and viral species/strains and in functional pathways observed in our analysis may influence microbiome engraftment in neonates, which may impact their future clinical outcomes.


Subject(s)
Celiac Disease/microbiology , Diet, Gluten-Free , Metabolome , Microbiota , Milk, Human/microbiology , Adult , Case-Control Studies , Celiac Disease/diet therapy , Cross-Sectional Studies , Female , Glutens/metabolism , Humans , Infant, Newborn , Metabolomics , Metagenomics , Prospective Studies
12.
Nutrients ; 13(7)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201435

ABSTRACT

To date, the only treatment for celiac disease (CD) consists of a strict lifelong gluten-free diet (GFD), which has numerous limitations in patients with CD. For this reason, dietary transgressions are frequent, implying intestinal damage and possible long-term complications. There is an unquestionable need for non-dietary alternatives to avoid damage by involuntary contamination or voluntary dietary transgressions. In recent years, different therapies and treatments for CD have been developed and studied based on the degradation of gluten in the intestinal lumen, regulation of the immune response, modulation of intestinal permeability, and induction of immunological tolerance. In this review, therapeutic lines for CD are evaluated with special emphasis on phase III and II clinical trials, some of which have promising results.


Subject(s)
Celiac Disease/therapy , Celiac Disease/immunology , Celiac Disease/microbiology , Clinical Trials as Topic , Diet, Gluten-Free , Gastrointestinal Microbiome , Glutens/adverse effects , Humans , Immunomodulation
13.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34253606

ABSTRACT

Other than exposure to gluten and genetic compatibility, the gut microbiome has been suggested to be involved in celiac disease (CD) pathogenesis by mediating interactions between gluten/environmental factors and the host immune system. However, to establish disease progression markers, it is essential to assess alterations in the gut microbiota before disease onset. Here, a prospective metagenomic analysis of the gut microbiota of infants at risk of CD was done to track shifts in the microbiota before CD development. We performed cross-sectional and longitudinal analyses of gut microbiota, functional pathways, and metabolites, starting from 18 mo before CD onset, in 10 infants who developed CD and 10 matched nonaffected infants. Cross-sectional analysis at CD onset identified altered abundance of six microbial strains and several metabolites between cases and controls but no change in microbial species or pathway abundance. Conversely, results of longitudinal analysis revealed several microbial species/strains/pathways/metabolites occurring in increased abundance and detected before CD onset. These had previously been linked to autoimmune and inflammatory conditions (e.g., Dialister invisus, Parabacteroides sp., Lachnospiraceae, tryptophan metabolism, and metabolites serine and threonine). Others occurred in decreased abundance before CD onset and are known to have anti-inflammatory effects (e.g., Streptococcus thermophilus, Faecalibacterium prausnitzii, and Clostridium clostridioforme). Additionally, we uncovered previously unreported microbes/pathways/metabolites (e.g., Porphyromonas sp., high mannose-type N-glycan biosynthesis, and serine) that point to CD-specific biomarkers. Our study establishes a road map for prospective longitudinal study designs to better understand the role of gut microbiota in disease pathogenesis and therapeutic targets to reestablish tolerance and/or prevent autoimmunity.


Subject(s)
Celiac Disease/microbiology , Gastrointestinal Microbiome , Autoimmunity , Biomarkers/metabolism , Celiac Disease/metabolism , Child, Preschool , Cross-Sectional Studies , Female , Gastrointestinal Microbiome/genetics , Host Microbial Interactions , Humans , Infant , Inflammation , Longitudinal Studies , Male , Metabolic Networks and Pathways , Metabolome , Metagenomics , Prospective Studies
14.
Front Immunol ; 12: 642166, 2021.
Article in English | MEDLINE | ID: mdl-34163468

ABSTRACT

The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.


Subject(s)
Celiac Disease/immunology , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases/immunology , Animals , Celiac Disease/microbiology , Child , Diabetes Mellitus, Type 1/microbiology , Female , Humans , Inflammatory Bowel Diseases/microbiology , Male
15.
Biosci Rep ; 41(6)2021 06 25.
Article in English | MEDLINE | ID: mdl-34076695

ABSTRACT

The microbiome plays an important role in maintaining human health. Despite multiple factors being attributed to the shaping of the human microbiome, extrinsic factors such diet and use of medications including antibiotics appear to dominate. Mucosal surfaces, particularly in the gut, are highly adapted to be able to tolerate a large population of microorganisms whilst still being able to produce a rapid and effective immune response against infection. The intestinal microbiome is not functionally independent from the host mucosa and can, through presentation of microbe-associated molecular patterns (MAMPs) and generation of microbe-derived metabolites, fundamentally influence mucosal barrier integrity and modulate host immunity. In a healthy gut there is an abundance of beneficial bacteria that help to preserve intestinal homoeostasis, promote protective immune responses, and limit excessive inflammation. The importance of the microbiome is further highlighted during dysbiosis where a loss of this finely balanced microbial population can lead to mucosal barrier dysfunction, aberrant immune responses, and chronic inflammation that increases the risk of disease development. Improvements in our understanding of the microbiome are providing opportunities to harness members of a healthy microbiota to help reverse dysbiosis, reduce inflammation, and ultimately prevent disease progression.


Subject(s)
Bacteria/metabolism , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Intestines/microbiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/microbiology , Autoimmune Diseases/therapy , Bacteria/immunology , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/microbiology , Celiac Disease/therapy , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/therapy , Humans , Immunity, Mucosal , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Intestines/immunology , Intestines/metabolism , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Metabolic Syndrome/therapy , Pathogen-Associated Molecular Pattern Molecules/metabolism , Probiotics/therapeutic use , Signal Transduction
16.
Methods Mol Biol ; 2325: 229-241, 2021.
Article in English | MEDLINE | ID: mdl-34053062

ABSTRACT

Gut microbiota, the largest microbial community living in the human body, exerts a variety of metabolic, structural, and functional actions. In particular, it is essential for the full immune system development and maturation, as demonstrated by studies on germ-free animals, showing immune impairment at different levels. Gut microbiota shapes the immune responses by promoting immune tolerance toward food antigens and commensals in the steady state. This process is orchestrated by a complex network of both microbial and human cells and molecular mediators. Microbiota eubiosis is fundamental in establishing a correct balance between tolerance and immunity. Contrarily, microbiota dysbiosis is correlated with alterations in the immune balance, as evidenced in intestinal pathologies characterized by aberrant immune responses, such as inflammatory bowel disease and celiac disease, in which either break of tolerance against commensals or microbial dysbiosis is reported. On the other hand, a role for gut microbiota in stimulating the cytotoxic immune response in contexts of immunosuppression, like the ones featuring tumors and vaccinations, is emerging. The bifaceted role of gut microbiota in the delicate balance between tolerance and immunity could be exploited in order to develop pioneering therapeutic strategies, complementary to the pharmacological ones, thus representing a field worthy of further studies specifically focused on this topic.


Subject(s)
Celiac Disease/microbiology , Gastrointestinal Microbiome/immunology , Immune Tolerance , Inflammatory Bowel Diseases/microbiology , Neoplasms/immunology , Neoplasms/microbiology , T-Lymphocytes, Cytotoxic/immunology , Celiac Disease/immunology , Celiac Disease/metabolism , Dendritic Cells/immunology , Dysbiosis/immunology , Dysbiosis/metabolism , Humans , Immune Checkpoint Inhibitors , Immunoglobulin A/immunology , Immunotherapy/methods , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology
17.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923593

ABSTRACT

In the last decade, the widespread application of shotgun metagenomics provided extensive characterization of the bacterial "dark matter" of the gut microbiome, propelling the development of dedicated, standardized bioinformatic pipelines and the systematic collection of metagenomic data into comprehensive databases. The advent of next-generation sequencing also unravels a previously underestimated viral population (virome) present in the human gut. Despite extensive efforts to characterize the human gut virome, to date, little is known about the childhood gut virome. However, alterations of the gut virome in children have been linked to pathological conditions such as inflammatory bowel disease, type 1 diabetes, malnutrition, diarrhea and celiac disease.


Subject(s)
Celiac Disease/virology , Diabetes Mellitus, Type 1/virology , Diarrhea/virology , Inflammatory Bowel Diseases/virology , Intestinal Mucosa/virology , Virome , Celiac Disease/microbiology , Child , Diabetes Mellitus, Type 1/microbiology , Diarrhea/microbiology , Humans , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Metagenome
18.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669290

ABSTRACT

The use of inorganic nanoparticles (NPs) has expanded into various industries including food manufacturing, agriculture, cosmetics, and construction. This has allowed NPs access to the human gastrointestinal tract, yet little is known about how they may impact human health. As the gut microbiome continues to be increasingly implicated in various diseases of unknown etiology, researchers have begun studying the potentially toxic effects of these NPs on the gut microbiome. Unfortunately, conflicting results have limited researcher's ability to evaluate the true impact of NPs on the gut microbiome in relation to health. This review focuses on the impact of five inorganic NPs (silver, iron oxide, zinc oxide, titanium dioxide, and silicon dioxide) on the gut microbiome and gastrointestinal tract with consideration for various methodological differences within the literature. This is important as NP-induced changes to the gut could lead to various gut-related diseases. These include irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), celiac disease, and colorectal cancer. Research in this area is necessary as the use of NPs in various industries continues to grow along with the number of people suffering from chronic gastrointestinal diseases.


Subject(s)
Ferric Compounds/adverse effects , Food Industry , Gastrointestinal Microbiome/drug effects , Metal Nanoparticles/adverse effects , Metal Nanoparticles/chemistry , Silicon Dioxide/adverse effects , Silver/adverse effects , Titanium/adverse effects , Zinc Oxide/adverse effects , Celiac Disease/chemically induced , Celiac Disease/microbiology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/microbiology , Dysbiosis/chemically induced , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/microbiology , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/microbiology
19.
Int Rev Cell Mol Biol ; 358: 1-45, 2021.
Article in English | MEDLINE | ID: mdl-33707051

ABSTRACT

Celiac Disease (CeD) is an immune-mediated complex disease that is triggered by the ingestion of gluten and develops in genetically susceptible individuals. It has been known for a long time that the Human Leucocyte Antigen (HLA) molecules DQ2 and DQ8 are necessary, although not sufficient, for the disease development, and therefore other susceptibility genes and (epi)genetic events must participate in CeD pathogenesis. The advances in Genomics during the last 15 years have made CeD one of the immune-related disorders with the best-characterized genetic component. In the present work, we will first review the main Genome-Wide Association Studies (GWAS) carried out in the disorder, and emphasize post-GWAS discoveries, including diverse integrative strategies, SNP prioritization approaches, and insights into the Microbiome through the host Genomics. Second, we will explore CeD-related Epigenetics and Epigenomics, mostly focusing on the emerging knowledge of the celiac methylome, and the vast but yet under-explored non-coding RNA (ncRNA) landscape. We conclude that much has been done in the field although there are still completely unvisited areas in the post-Genomics of CeD. Chromatin conformation and accessibility, and Epitranscriptomics are promising domains that need to be unveiled to complete the big picture of the celiac Genome.


Subject(s)
Celiac Disease/genetics , Genetic Predisposition to Disease , Genome , Animals , Celiac Disease/microbiology , DNA Methylation/genetics , Genomics , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
20.
Dig Liver Dis ; 53(8): 972-979, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33741248

ABSTRACT

BACKGROUND: Duodenal dysbiosis has been suggested to possibly influence the clinical manifestations of coeliac disease (CD), both at onset and when symptoms persist despite a gluten-free diet (GFD). AIMS: To evaluate the relationship between duodenal microbiota composition and: i) clinical phenotype of untreated CD (UCD); ii) presence and type of persistent symptoms despite a satisfactory serological and histological response to a strict GFD. METHODS: Duodenal microbiota was analyzed by 16S rRNA sequencing and compared with i) clinical features in 12 adult UCD patients; ii) presence/absence and type of persistent symptoms (diarrhea-predominant vs. non-diarrhea predominant) in 25 adult treated coeliac patients (TCD) on a strict GFD. RESULTS: UCD with iron deficiency anemia (IDA) had a pro-inflammatory shift in their duodenal microbiota (reduction of Firmicutes, p = 0.03; increase of beta-Proteobacteria, p = 0.02) than those without IDA. TCD with persistent diarrhea showed a reduction of Actinobacteria (p = 0.03) and Rothia spp (p = 0.046) compared to TCD suffering from other type of persistent symptoms. CONCLUSION: A distinctive duodenal microbiota profile is associated with IDA in UCD, and diarrhea-predominant persistent symptoms in TCD. Clinical interventions may include reconsidering patients presenting with IDA as a specific disease subtype, and dietary rebalancing if diarrhea persists despite histological response to a GFD.


Subject(s)
Anemia, Iron-Deficiency/microbiology , Celiac Disease/microbiology , Diarrhea/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/genetics , Adult , Anemia, Iron-Deficiency/pathology , Celiac Disease/diet therapy , Celiac Disease/pathology , Diarrhea/pathology , Diet, Gluten-Free , Duodenum/microbiology , Dysbiosis/pathology , Feces/microbiology , Female , Humans , Male , Middle Aged , Prospective Studies , RNA, Ribosomal, 16S/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...