Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 283
Filtrar
1.
Sci Rep ; 14(1): 16712, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39030381

RESUMEN

The association between oral dysbiosis and celiac disease (CD) remains poorly understood, as does the impact of CD-associated dysbiosis on disease development or exacerbation. This study aims to investigate alterations in salivary microbial composition among children with CD. In this cross-sectional study, saliva samples from 12 children with active CD (A-CD group), 14 children with CD on a gluten-free diet (GFD), and 10 healthy control (HC) children were analyzed using DNA sequencing targeting the 16S ribosomal RNA. Both patients in A-CD and GFD groups showed a significant increase (p = 0.0001) in the Bacteroidetes phylum, while the Actinobacteria phylum showed a significant decrease (p = 0.0001). Notably, the Rothia genus and R.aeria also demonstrated a significant decrease (p = 0.0001) within the both CD groups as compare to HC. Additionally, the control group displayed a significant increase (p = 0.006) in R.mucilaginosa species compared to both CD patient groups. Distinct bacterial strains were abundant in the saliva of patients with active CD, indicating a unique composition of the salivary microbiome in individuals with CD. These findings suggest that our approach to assessing salivary microbiota changes may contribute to developing noninvasive methods for diagnosing and treating CD.


Asunto(s)
Enfermedad Celíaca , Microbiota , ARN Ribosómico 16S , Saliva , Humanos , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/diagnóstico , Saliva/microbiología , Niño , Femenino , Masculino , Estudios Transversales , ARN Ribosómico 16S/genética , Dieta Sin Gluten , Adolescente , Preescolar , Disbiosis/microbiología , Disbiosis/diagnóstico , Estudios de Casos y Controles , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación
2.
Adv Exp Med Biol ; 1449: 95-111, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39060733

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Probióticos , Humanos , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/genética , Enfermedad Celíaca/prevención & control , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/prevención & control , Probióticos/uso terapéutico
3.
World J Gastroenterol ; 30(24): 3044-3047, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38983964

RESUMEN

We comment here on the article by Stefanolo et al entitled "Effect of Aspergillus niger prolyl endopeptidase in patients with celiac disease on a long-term gluten-free diet", published in the World Journal of Gastroenterology. Celiac disease is a well-recognized systemic autoimmune disorder. In genetically susceptible people, the most evident damage is located in the small intestine, and is caused and worsened by the ingestion of gluten. For that reason, celiac patients adopt a gluten-free diet (GFD), but it has some limitations, and it does not prevent re-exposure to gluten. Research aims to develop adjuvant therapies, and one of the most studied alternatives is supplementation with Aspergillus niger prolyl endopeptidase protease (AN-PEP), which is able to degrade gluten in the stomach, reducing its concentration in the small intestine. The study found a high adherence to the GFD, but did not address AN-PEP as a gluten immunogenic peptide reducer, as it was only tested in patients following a GFD and not in gluten-exposing conditions. This study opens up new research perspectives in this area and shows that further study is needed to clarify the points that are still in doubt.


Asunto(s)
Aspergillus niger , Enfermedad Celíaca , Dieta Sin Gluten , Glútenes , Prolil Oligopeptidasas , Serina Endopeptidasas , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/enzimología , Humanos , Aspergillus niger/enzimología , Serina Endopeptidasas/metabolismo , Glútenes/inmunología , Glútenes/metabolismo , Glútenes/efectos adversos , Intestino Delgado/microbiología , Intestino Delgado/enzimología , Resultado del Tratamiento
4.
Microbiol Spectr ; 12(7): e0352423, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38860826

RESUMEN

Gluten possesses unique properties that render it only partially digestible. Consequently, it exerts detrimental effects on a part of the worldwide population who are afflicted with celiac disease (1%) or related disorders (5%), particularly due to the potential for cross-contamination even when adhering to a gluten-free diet (GFD). Finding solutions to break down gluten during digestion has a high nutritional and social impact. Here, a randomized double-blind placebo-controlled in vivo challenge investigated the gluten-degrading activity of a novel probiotic preparation comprising lactobacilli and their cytoplasmic extracts, Bacillus sp., and bacterial protease. In our clinical trial, we collected feces from 70 healthy volunteers at specific time intervals. Probiotic/placebo administration lasted 32 days, followed by 10 days of wash-out. After preliminary GFD to eliminate residual gluten from feces, increasing amounts of gluten (50 mg-10 g) were administered, each one for 4 consecutive days. Compared to placebo, the feces of volunteers fed with probiotics showed much lower amounts of residual gluten, mainly with increased intakes. Probiotics also regulate the intestinal microbial communities, improving the abundance of genera pivotal to maintaining homeostasis. Quantitative PCR confirmed that all probiotics persisted during the intervention, some also during wash-out. Probiotics promoted a fecal metabolome with potential immunomodulating activity, mainly related to derivatives of branched-chain amino acids and short-chain fatty acids. IMPORTANCE: The untapped potential of gluten-degrading bacteria and their application in addressing the recognized limitations of gluten-related disorder management and the ongoing risk of cross-contamination even when people follow a gluten-free diet (GFD) emphasizes the significance of the work. Because gluten, a common protein found in many cereals, must be strictly avoided to stop autoimmune reactions and related health problems, celiac disease and gluten sensitivity present difficult hurdles. However, because of the hidden presence of gluten in many food products and the constant danger of cross-contamination during food preparation and processing, total avoidance is frequently challenging. Our study presents a novel probiotic preparation suitable for people suffering from gluten-related disorders during GFD and for healthy individuals because it enhances gluten digestion and promotes gut microbiota functionality.


Asunto(s)
Heces , Microbioma Gastrointestinal , Glútenes , Probióticos , Humanos , Probióticos/administración & dosificación , Glútenes/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Heces/microbiología , Heces/química , Método Doble Ciego , Adulto , Masculino , Femenino , Lactobacillus/metabolismo , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/dietoterapia , Dieta Sin Gluten , Bacillus/metabolismo , Persona de Mediana Edad , Adulto Joven
5.
Nutrients ; 16(11)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38892582

RESUMEN

There is increasing evidence indicating that changes in both the composition and functionality of the intestinal microbiome are closely associated with the development of several chronic inflammatory diseases, with celiac disease (CeD) being particularly noteworthy. Thanks to the advent of culture-independent methodologies, the ability to identify and quantify the diverse microbial communities residing within the human body has been significantly improved. However, in the context of CeD, a notable challenge lies in characterizing the specific microbiota present on the mucosal surfaces of the intestine, rather than relying solely on fecal samples, which may not fully represent the relevant microbial populations. Currently, our comprehension of the composition and functional importance of mucosa-associated microbiota (MAM) in CeD remains an ongoing field of research because the limited number of available studies have reported few and sometimes contradictory results. MAM plays a crucial role in the development and progression of CeD, potentially acting as both a trigger and modulator of the immune response within the intestinal mucosa, given its proximity to the epithelial cells and direct interaction. According to this background, this review aims to consolidate the existing literature specifically focused on MAM in CeD. By elucidating the complex interplay between the host immune system and the gut microbiota, we aim to pave the way for new interventions based on novel therapeutic targets and diagnostic biomarkers for MAM in CeD.


Asunto(s)
Enfermedad Celíaca , Duodeno , Microbioma Gastrointestinal , Mucosa Intestinal , Enfermedad Celíaca/microbiología , Humanos , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/microbiología , Duodeno/microbiología
6.
Nutrients ; 16(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38931237

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Disbiosis , Microbioma Gastrointestinal , Glútenes , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Humanos , Glútenes/inmunología , Glútenes/efectos adversos , Disbiosis/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo
7.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719750

RESUMEN

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.


Asunto(s)
Candida albicans , Enfermedad Celíaca , Homeostasis , Mastocitos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/metabolismo , Humanos , Candida albicans/patogenicidad , Candida albicans/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Microbioma Gastrointestinal/inmunología , Disbiosis/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Animales , Candida/patogenicidad , Candida/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
8.
Benef Microbes ; 15(2): 165-177, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38653476

RESUMEN

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.


Asunto(s)
Bacterias , Biomarcadores , Enfermedad Celíaca , Duodeno , Microbioma Gastrointestinal , ARN Ribosómico 16S , Estómago , Humanos , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/diagnóstico , Niño , Proyectos Piloto , Preescolar , Duodeno/microbiología , Duodeno/patología , Adolescente , Masculino , ARN Ribosómico 16S/genética , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estómago/microbiología , Estómago/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Biopsia , ADN Bacteriano/genética
9.
Virulence ; 15(1): 2334085, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38528835

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Enfermedad de Crohn , Humanos , Candida albicans/fisiología , Enfermedad Celíaca/microbiología , Anticuerpos Antifúngicos , Formación de Anticuerpos
10.
Gastroenterology ; 167(1): 34-50, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38286392

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , Índice de Severidad de la Enfermedad , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/genética , Humanos , Animales , Factores de Riesgo , Predisposición Genética a la Enfermedad , Glútenes/inmunología , Glútenes/efectos adversos , Dieta/efectos adversos , Modelos Animales de Enfermedad
11.
Gut Microbes ; 15(1): 2172955, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36751856

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , MicroARNs , Adulto , Humanos , Niño , Enfermedad Celíaca/microbiología , Dieta Sin Gluten , Glútenes/efectos adversos
12.
Clin Exp Med ; 23(6): 2773-2788, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36609792

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , Probióticos , Humanos , Enfermedad Celíaca/terapia , Enfermedad Celíaca/microbiología , Probióticos/uso terapéutico , Dieta Sin Gluten , Disbiosis/terapia
13.
J Gastroenterol Hepatol ; 37(10): 1844-1852, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35734803

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Antibacterianos/uso terapéutico , Pruebas Respiratorias , Estudios de Casos y Controles , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/microbiología , Humanos , Intestino Delgado/microbiología , Prevalencia
14.
Sci Rep ; 12(1): 6607, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35459889

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Limosilactobacillus reuteri , Microbiota , Bacterias/genética , Enfermedad Celíaca/microbiología , Niño , Preescolar , Femenino , Humanos , Receptores de Lipopolisacáridos , Leche Humana/microbiología
15.
Dig Dis Sci ; 67(1): 216-223, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33723701

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Disbiosis , Hongos , Micobioma , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/fisiopatología , Niño , Disbiosis/diagnóstico , Disbiosis/microbiología , Heces/microbiología , Femenino , Hongos/clasificación , Hongos/inmunología , Hongos/aislamiento & purificación , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Metagenómica/métodos , Fenómenos Microbiológicos , Micobioma/genética , Micobioma/inmunología , Arabia Saudita/epidemiología
16.
Am J Surg Pathol ; 46(1): e43-e54, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33739793

RESUMEN

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.


Asunto(s)
Autoinmunidad , Enfermedad Celíaca/inmunología , Duodeno/inmunología , Ambiente , Subgrupos de Linfocitos T/inmunología , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Biopsia , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/patología , Enfermedad Celíaca/terapia , Duodeno/microbiología , Duodeno/patología , Disbiosis , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Humanos , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Medición de Riesgo , Factores de Riesgo
17.
Gastroenterology ; 161(5): 1395-1411.e4, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34416277

RESUMEN

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.


Asunto(s)
Bacterias/inmunología , Enfermedad Celíaca/terapia , Microbioma Gastrointestinal , Glútenes/inmunología , Fenómenos Inmunogenéticos , Pruebas Inmunológicas , Intestinos/inmunología , Animales , Bacterias/patogenicidad , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Intestinos/patología , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo
18.
Nutrients ; 13(7)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201435

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca/terapia , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Ensayos Clínicos como Asunto , Dieta Sin Gluten , Microbioma Gastrointestinal , Glútenes/efectos adversos , Humanos , Inmunomodulación
19.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34253606

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca/microbiología , Microbioma Gastrointestinal , Autoinmunidad , Biomarcadores/metabolismo , Enfermedad Celíaca/metabolismo , Preescolar , Estudios Transversales , Femenino , Microbioma Gastrointestinal/genética , Interacciones Microbiota-Huesped , Humanos , Lactante , Inflamación , Estudios Longitudinales , Masculino , Redes y Vías Metabólicas , Metaboloma , Metagenómica , Estudios Prospectivos
20.
Nutrients ; 13(7)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34210038

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca/microbiología , Dieta Sin Gluten , Metaboloma , Microbiota , Leche Humana/microbiología , Adulto , Estudios de Casos y Controles , Enfermedad Celíaca/dietoterapia , Estudios Transversales , Femenino , Glútenes/metabolismo , Humanos , Recién Nacido , Metabolómica , Metagenómica , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA