Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36591617

RESUMEN

OBJECTIVE: Dietary therapies for irritable bowel syndrome (IBS) have received increasing interest but predicting which patients will benefit remains a challenge due to a lack of mechanistic insight. We recently found evidence of a role for the microbiota in dietary modulation of pain signalling in a humanised mouse model of IBS. This randomised cross-over study aimed to test the hypothesis that pain relief following reduced consumption of fermentable carbohydrates is the result of changes in luminal neuroactive metabolites. DESIGN: IBS (Rome IV) participants underwent four trial periods: two non-intervention periods, followed by a diet low (LFD) and high in fermentable carbohydrates for 3 weeks each. At the end of each period, participants completed questionnaires and provided stool. The effects of faecal supernatants (FS) collected before (IBS FS) and after a LFD (LFD FS) on nociceptive afferent neurons were assessed in mice using patch-clamp and ex vivo colonic afferent nerve recording techniques. RESULTS: Total IBS symptom severity score and abdominal pain were reduced by the LFD (N=25; p<0.01). Excitability of neurons was increased in response to IBS FS, but this effect was reduced (p<0.01) with LFD FS from pain-responders. IBS FS from pain-responders increased mechanosensitivity of nociceptive afferent nerve axons (p<0.001), an effect lost following LFD FS administration (p=NS) or when IBS FS was administered in the presence of antagonists of histamine receptors or protease inhibitors. CONCLUSIONS: In a subset of IBS patients with improvement in abdominal pain following a LFD, there is a decrease in pronociceptive signalling from FS, suggesting that changes in luminal mediators may contribute to symptom response.

2.
Clin Gastroenterol Hepatol ; 19(11): 2343-2352.e8, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-32827724

RESUMEN

BACKGROUND & AIMS: Many patients with irritable bowel syndrome (IBS) perceive that their symptoms are triggered by wheat-containing foods. We assessed symptoms and gastrointestinal transit before and after a gluten-free diet (GFD) in unselected patients with IBS and investigated biomarkers associated with symptoms. METHODS: We performed a prospective study of 50 patients with IBS (ROME III, all subtypes), with and without serologic reactivity to gluten (antigliadin IgG and IgA), and 25 healthy subjects (controls) at a university hospital in Hamilton, Ontario, Canada, between 2012 and 2016. Gastrointestinal transit, gut symptoms, anxiety, depression, somatization, dietary habits, and microbiota composition were studied before and after 4 weeks of a GFD. HLA-DQ2/DQ8 status was determined. GFD compliance was assessed by a dietitian and by measuring gluten peptides in stool. RESULTS: There was no difference in symptoms among patients at baseline, but after the GFD, patients with antigliadin IgG and IgA reported less diarrhea than patients without these antibodies (P = .03). Compared with baseline, IBS symptoms improved in 18 of 24 patients (75%) with antigliadin IgG and IgA and in 8 of 21 patients (38%) without the antibodies. Although constipation, diarrhea, and abdominal pain were reduced in patients with antigliadin IgG and IgA, only pain decreased in patients without these antibodies. Gastrointestinal transit normalized in a higher proportion of patients with antigliadin IgG and IgA. Anxiety, depression, somatization, and well-being increased in both groups. The presence of antigliadin IgG was associated with overall reductions in symptoms (adjusted odds ratio compared with patients without this antibody, 128.9; 95% CI, 1.16-1427.8; P = .04). Symptoms were reduced even in patients with antigliadin IgG and IgA who reduced gluten intake but were not strictly compliant with the GFD. In controls, a GFD had no effect on gastrointestinal symptoms or gut function. CONCLUSIONS: Antigliadin IgG can be used as a biomarker to identify patients with IBS who might have reductions in symptoms, particularly diarrhea, on a GFD. Larger studies are needed to validate these findings. ClinicalTrials.gov: NCT03492333.


Asunto(s)
Enfermedad Celíaca , Síndrome del Colon Irritable , Diarrea , Dieta Sin Gluten , Humanos , Inmunoglobulina G , Estudios Prospectivos
3.
Neuropsychobiology ; 79(1): 5-12, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30928978

RESUMEN

BACKGROUND: Emerging evidence suggests an important role of the human gut microbiome in psychiatry and neurodevelopmental disorders. An increasing body of literature based on animal studies has reported that the gut microbiome influences brain development and behavior by interacting with the gut-brain axis. Furthermore, as the gut microbiome has an important role in metabolism and is known to interact with pharmaceuticals, recent evidence suggests a role for the microbiome in antipsychotic-induced metabolic side effects in animals and humans. PURPOSE: Here we present the protocol for a two-phase study investigating the gut microbiome in healthy controls and in patients with schizophrenia treated with antipsychotics. METHODS: Phase I of our study involves humans exclusively. We recruit 25 patients who are chronically treated with clozapine and compare them with 25 healthy controls matched for age, sex, BMI, and smoking status. A second cohort consists of 25 patients newly starting on clozapine, and a third cohort includes 25 antipsychotic-naive patients. The patients in the second cohort and third cohort are prospectively assessed for up to 6 and 12 weeks, respectively. Phase II of this study will incorporate microbiota humanized mouse models to examine the influence of human fecal transplant on metabolic parameters and the gut-brain axis. Progress and Future Directions: We are underway with the first participants enrolled in all phase I treatment cohorts. This study will contribute to elucidating the role of the gut microbiome in schizophrenia and metabolic side effects. In addition, its results may help to explore potential therapeutic targets for antipsychotic-induced metabolic side effects.


Asunto(s)
Antipsicóticos/efectos adversos , Clozapina/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/microbiología , Aumento de Peso/efectos de los fármacos , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Estudios Prospectivos
4.
Gastroenterology ; 153(2): 448-459.e8, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28483500

RESUMEN

BACKGROUND & AIMS: Probiotics can reduce symptoms of irritable bowel syndrome (IBS), but little is known about their effects on psychiatric comorbidities. We performed a prospective study to evaluate the effects of Bifidobacterium longum NCC3001 (BL) on anxiety and depression in patients with IBS. METHODS: We performed a randomized, double-blind, placebo-controlled study of 44 adults with IBS and diarrhea or a mixed-stool pattern (based on Rome III criteria) and mild to moderate anxiety and/or depression (based on the Hospital Anxiety and Depression scale) at McMaster University in Canada, from March 2011 to May 2014. At the screening visit, clinical history and symptoms were assessed and blood samples were collected. Patients were then randomly assigned to groups and given daily BL (n = 22) or placebo (n = 22) for 6 weeks. At weeks 0, 6, and 10, we determined patients' levels of anxiety and depression, IBS symptoms, quality of life, and somatization using validated questionnaires. At weeks 0 and 6, stool, urine and blood samples were collected, and functional magnetic resonance imaging (fMRI) test was performed. We assessed brain activation patterns, fecal microbiota, urine metabolome profiles, serum markers of inflammation, neurotransmitters, and neurotrophin levels. RESULTS: At week 6, 14 of 22 patients in the BL group had reduction in depression scores of 2 points or more on the Hospital Anxiety and Depression scale, vs 7 of 22 patients in the placebo group (P = .04). BL had no significant effect on anxiety or IBS symptoms. Patients in the BL group had a mean increase in quality of life score compared with the placebo group. The fMRI analysis showed that BL reduced responses to negative emotional stimuli in multiple brain areas, including amygdala and fronto-limbic regions, compared with placebo. The groups had similar fecal microbiota profiles, serum markers of inflammation, and levels of neurotrophins and neurotransmitters, but the BL group had reduced urine levels of methylamines and aromatic amino acids metabolites. At week 10, depression scores were reduced in patients given BL vs placebo. CONCLUSION: In a placebo-controlled trial, we found that the probiotic BL reduces depression but not anxiety scores and increases quality of life in patients with IBS. These improvements were associated with changes in brain activation patterns that indicate that this probiotic reduces limbic reactivity. ClinicalTrials.gov no. NCT01276626.


Asunto(s)
Bifidobacterium longum , Encéfalo/fisiopatología , Depresión/terapia , Síndrome del Colon Irritable/psicología , Probióticos/administración & dosificación , Adulto , Ansiedad/fisiopatología , Ansiedad/psicología , Ansiedad/terapia , Encéfalo/diagnóstico por imagen , Encéfalo/microbiología , Canadá , Depresión/fisiopatología , Depresión/psicología , Diarrea/microbiología , Diarrea/terapia , Método Doble Ciego , Emociones , Heces/microbiología , Femenino , Humanos , Síndrome del Colon Irritable/fisiopatología , Síndrome del Colon Irritable/terapia , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Calidad de Vida , Encuestas y Cuestionarios , Resultado del Tratamiento
5.
Gut ; 66(7): 1241-1251, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-26976734

RESUMEN

OBJECTIVE: To gain mechanistic insights, we compared effects of low fermentable oligosaccharides, disaccharides and monosaccharides and polyols (FODMAP) and high FODMAP diets on symptoms, the metabolome and the microbiome of patients with IBS. DESIGN: We performed a controlled, single blind study of patients with IBS (Rome III criteria) randomised to a low (n=20) or high (n=20) FODMAP diet for 3 weeks. Symptoms were assessed using the IBS symptom severity scoring (IBS-SSS). The metabolome was evaluated using the lactulose breath test (LBT) and metabolic profiling in urine using mass spectrometry. Stool microbiota composition was analysed by 16S rRNA gene profiling. RESULTS: Thirty-seven patients (19 low FODMAP; 18 high FODMAP) completed the 3-week diet. The IBS-SSS was reduced in the low FODMAP diet group (p<0.001) but not the high FODMAP group. LBTs showed a minor decrease in H2 production in the low FODMAP compared with the high FODMAP group. Metabolic profiling of urine showed groups of patients with IBS differed significantly after the diet (p<0.01), with three metabolites (histamine, p-hydroxybenzoic acid, azelaic acid) being primarily responsible for discrimination between the two groups. Histamine, a measure of immune activation, was reduced eightfold in the low FODMAP group (p<0.05). Low FODMAP diet increased Actinobacteria richness and diversity, and high FODMAP diet decreased the relative abundance of bacteria involved in gas consumption. CONCLUSIONS: IBS symptoms are linked to FODMAP content and associated with alterations in the metabolome. In subsets of patients, FODMAPs modulate histamine levels and the microbiota, both of which could alter symptoms. TRIAL REGISTRATION NUMBER: NCT01829932.


Asunto(s)
Dieta , Síndrome del Colon Irritable/dietoterapia , Adulto , Anciano , Anciano de 80 o más Años , Pruebas Respiratorias , Ácidos Dicarboxílicos/orina , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Histamina/orina , Humanos , Síndrome del Colon Irritable/metabolismo , Lactulosa , Masculino , Metaboloma , Persona de Mediana Edad , Parabenos/análisis , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Método Simple Ciego
7.
J Cell Physiol ; 231(11): 2529-40, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27100271

RESUMEN

Polymorphisms in the PTPN11 gene encoding for the tyrosine phosphatase SHP-2 were described in patients with ulcerative colitis. We have recently demonstrated that mice with an intestinal epithelial cell-specific deletion of SHP-2 (SHP-2(IEC-KO) ) develop severe colitis 1 month after birth. However, the mechanisms by which SHP-2 deletion induces colonic inflammation remain to be elucidated. We generated SHP-2(IEC-KO) mice lacking Myd88 exclusively in the intestinal epithelium. The colonic phenotype was histologically analyzed and cell differentiation was determined by electron microscopy and lysozyme or Alcian blue staining. Microbiota composition was analyzed by 16S sequencing. Results show that innate defense genes including those specific to Paneth cells were strongly up-regulated in SHP-2-deficient colons. Expansion of intermediate cells (common progenitors of the Goblet and Paneth cell lineages) was found in the colon of SHP-2(IEC-KO) mice whereas Goblet cell number was clearly diminished. These alterations in Goblet/intermediate cell ratio were noticed 2 weeks after birth, before the onset of inflammation and were associated with significant alterations in microbiota composition. Indeed, an increase in Enterobacteriaceae and a decrease in Firmicutes were observed in the colon of these mice, indicating that dysbiosis also occurred prior to inflammation. Importantly, loss of epithelial Myd88 expression inhibited colitis development in SHP-2(IEC-KO) mice, rescued Goblet/intermediate cell ratio, and prevented NFκB hyperactivation and inflammation. These data indicate that SHP-2 is functionally important for the maintenance of appropriate barrier function and host-microbiota homeostasis in the large intestine. J. Cell. Physiol. 231: 2529-2540, 2016. © 2016 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular , Colon/patología , Homeostasis , Inflamación/patología , Inflamación/prevención & control , Microbiota , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Animales Recién Nacidos , Péptidos Catiónicos Antimicrobianos/metabolismo , Biomarcadores/metabolismo , Células Caliciformes/metabolismo , Células Caliciformes/patología , Inflamación/genética , Ratones Endogámicos C57BL , Muramidasa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Regulación hacia Arriba/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 311(5): G920-G933, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27514477

RESUMEN

The cholinergic anti-inflammatory pathway reduces systemic tumor necrosis factor (TNF) via acetylcholine-producing memory T cells in the spleen. These choline acetyltransferase (ChAT)-expressing T cells are also found in the intestine, where their function is unclear. We aimed to characterize these cells in mouse and human intestine and delineate their function. We made use of the ChAT-enhanced green fluorescent protein (eGFP) reporter mice. CD4Cre mice were crossed to ChATfl/fl mice to achieve specific deletion of ChAT in CD4+ T cells. We observed that the majority of ChAT-expressing T cells in the human and mouse intestine have characteristics of Th17 cells and coexpress IL17A, IL22, and RORC The generation of ChAT-expressing T cells was skewed by dendritic cells after activation of their adrenergic receptor ß2 To evaluate ChAT T cell function, we generated CD4-specific ChAT-deficient mice. CD4ChAT-/- mice showed a reduced level of epithelial antimicrobial peptides lysozyme, defensin A, and ang4, which was associated with an enhanced bacterial diversity and richness in the small intestinal lumen in CD4ChAT-/- mice. We conclude that ChAT-expressing T cells in the gut are stimulated by adrenergic receptor activation on dendritic cells. ChAT-expressing T cells may function to mediate the host AMP secretion, microbial growth and expansion.


Asunto(s)
Acetilcolina/metabolismo , Defensinas/metabolismo , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Muramidasa/metabolismo , Ribonucleasa Pancreática/metabolismo , Linfocitos T/metabolismo , Animales , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos
9.
Am J Physiol Gastrointest Liver Physiol ; 308(12): G994-1003, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882612

RESUMEN

The small intestine is a significant site of ulceration and bleeding induced by nonsteroidal anti-inflammatory drugs (NSAIDs). The pathogenesis is poorly understood. The present study explored the roles of bile, bacteria, and enterohepatic circulation to NSAID enteropathy, using both a conventional NSAID (naproxen) and a gastrointestinal-safe naproxen derivative (ATB-346), as well as proton pump inhibitors (PPIs). Rats were treated orally with naproxen or equimolar doses of ATB-346 over a 5-day period, with or without PPI administration, and intestinal damage was quantified. The cytotoxicity of bile from the rats was evaluated in vitro. Biliary excretion of naproxen and ATB-346 was determined. The impact of the NSAIDs and of PPIs on the composition of the intestinal microbiota was examined by deep sequencing of 16s rRNA. Naproxen caused significant intestinal damage and inflammation, whereas ATB-346 did not. Naproxen, but not ATB-346, dose dependently increased the cytotoxicity of bile, and it was further increased by PPI coadministration. Whereas biliary excretion of naproxen was significant in naproxen-treated rats, it was greatly reduced in rats treated with ATB-346. The enteric microbiota of naproxen-treated rats was distinct from that in vehicle- or ATB-346-treated rats, and PPI administration caused significant intestinal dysbiosis. The increase in cytotoxicity of bile induced by naproxen and PPIs may contribute significantly to intestinal ulceration and bleeding. Some of these effects may occur secondary to significant changes in the jejunal microbiota induced by both naproxen and PPIs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Sulfuro de Hidrógeno/farmacología , Enfermedades Intestinales/tratamiento farmacológico , Naproxeno/análogos & derivados , Inhibidores de la Bomba de Protones/farmacología , Animales , Eliminación Hepatobiliar/fisiología , Inflamación/tratamiento farmacológico , Masculino , Naproxeno/farmacología , ARN Ribosómico 16S/genética , Ratas Wistar
11.
Eur J Nutr ; 54(1): 119-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24700375

RESUMEN

PURPOSE: To investigate whether breast-milk composition and microbiota differ in healthy mothers and mothers with celiac disease (CD) to ultimately contribute to identify additional factors determining CD risk. METHODS: Breast-milk samples from healthy mothers (n = 12) and mothers with CD (n = 12) were collected. Cytokines and secretory immunoglobulin A (sIgA) were analyzed by bead-arrays and flow cytometry and human milk oligosaccharides (HMOs) were assessed by capillary electrophoresis with laser-induced fluorescence (CE-LIF) detection. Breast-milk microbiota composition was analyzed by conventional and quantitative real-time PCR. RESULT: Breast milk from CD mothers showed significantly lower levels of interleukin (IL) 12p70 (P < 0.042), transforming growth factor (TGF)-ß1 (P < 0.018) and sIgA (P < 0.003) and almost significantly lower levels of interferon (IFN)-γ (P < 0.058). Six mothers in each group belonged to the secretor Le(a-b+) type, one to the secretor Le(a-b-) type and five to the non-secretor Le(a+b-) type. CD mothers of non-secretor Le(a+b-) type showed increased Lacto-N-tetraose content (P < 0.042) compared with healthy mothers. CD mothers' milk showed reduced gene copy numbers of Bifidobacterium spp. (P < 0.026) and B. fragilis group (P < 0.044). CONCLUSION: CD mothers' breast milk is characterized by a reduced abundance of immunoprotective compounds (TGF-ß1 and sIgA) and bifidobacteria. The reduction in these components could theoretically diminish the protective effects of breast-feeding on the child's future risk of developing CD.


Asunto(s)
Bacteroides fragilis/aislamiento & purificación , Bifidobacterium/aislamiento & purificación , Enfermedad Celíaca/metabolismo , Citocinas/análisis , Inmunoglobulina A Secretora/análisis , Leche Humana/química , Oligosacáridos/análisis , Adulto , Bacteroides fragilis/clasificación , Bacteroides fragilis/genética , Bacteroides fragilis/crecimiento & desarrollo , Bifidobacterium/clasificación , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Estudios de Casos y Controles , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Citocinas/metabolismo , Dieta Sin Gluten , Salud de la Familia , Femenino , Dosificación de Gen , Genes Bacterianos , Humanos , Inmunoglobulina A Secretora/metabolismo , Interferón gamma/análisis , Interferón gamma/metabolismo , Interleucina-12/análisis , Interleucina-12/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Leche Humana/microbiología , Tipificación Molecular , Oligosacáridos/metabolismo , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/metabolismo
12.
J Physiol ; 592(14): 2989-97, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24756641

RESUMEN

The gut-brain axis is the bidirectional communication between the gut and the brain, which occurs through multiple pathways that include hormonal, neural and immune mediators. The signals along this axis can originate in the gut, the brain or both, with the objective of maintaining normal gut function and appropriate behaviour. In recent years, the study of gut microbiota has become one of the most important areas in biomedical research. Attention has focused on the role of gut microbiota in determining normal gut physiology and immunity and, more recently, on its role as modulator of host behaviour ('microbiota-gut-brain axis'). We therefore review the literature on the role of gut microbiota in gut homeostasis and link it with mechanisms that could influence behaviour. We discuss the association of dysbiosis with disease, with particular focus on functional bowel disorders and their relationship to psychological stress. This is of particular interest because exposure to stressors has long been known to increase susceptibility to and severity of gastrointestinal diseases.


Asunto(s)
Encéfalo/fisiología , Enfermedades Gastrointestinales , Tracto Gastrointestinal , Microbiota , Estrés Psicológico , Animales , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/fisiología , Humanos , Estrés Psicológico/microbiología , Estrés Psicológico/fisiopatología
13.
Gut Microbes ; 16(1): 2360233, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38949979

RESUMEN

Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Enfermedades Gastrointestinales , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Humanos , Eje Cerebro-Intestino/fisiología , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Encéfalo/microbiología , Encéfalo/fisiopatología , Animales , Tracto Gastrointestinal/microbiología
14.
Cell Mol Gastroenterol Hepatol ; 17(3): 383-398, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38061549

RESUMEN

BACKGROUND & AIMS: Although chronic diarrhea and constipation are common, the treatment is symptomatic because their pathophysiology is poorly understood. Accumulating evidence suggests that the microbiota modulates gut function, but the underlying mechanisms are unknown. We therefore investigated the pathways by which microbiota modulates gastrointestinal motility in different sections of the alimentary tract. METHODS: Gastric emptying, intestinal transit, muscle contractility, acetylcholine release, gene expression, and vasoactive intestinal polypeptide (VIP) immunoreactivity were assessed in wild-type and Myd88-/-Trif-/- mice in germ-free, gnotobiotic, and specific pathogen-free conditions. Effects of transient colonization and antimicrobials as well as immune cell blockade were investigated. VIP levels were assessed in human full-thickness biopsies by Western blot. RESULTS: Germ-free mice had similar gastric emptying but slower intestinal transit compared with specific pathogen-free mice or mice monocolonized with Lactobacillus rhamnosus or Escherichia coli, the latter having stronger effects. Although muscle contractility was unaffected, its neural control was modulated by microbiota by up-regulating jejunal VIP, which co-localized with and controlled cholinergic nerve function. This process was responsive to changes in the microbial composition and load and mediated through toll-like receptor signaling, with enteric glia cells playing a key role. Jejunal VIP was lower in patients with chronic intestinal pseudo-obstruction compared with control subjects. CONCLUSIONS: Microbial control of gastrointestinal motility is both region- and bacteria-specific; it reacts to environmental changes and is mediated by innate immunity-neural system interactions. By regulating cholinergic nerves, small intestinal VIP plays a key role in this process, thus providing a new therapeutic target for patients with motility disorders.


Asunto(s)
Motilidad Gastrointestinal , Péptido Intestinal Vasoactivo , Humanos , Ratones , Animales , Péptido Intestinal Vasoactivo/metabolismo , Motilidad Gastrointestinal/fisiología , Neuroglía/metabolismo , Colinérgicos
15.
Eur J Nutr ; 52(2): 637-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22576041

RESUMEN

PURPOSE: In addition to genetic risk, environmental factors might influence coeliac disease (CD) development. We sought to assess the effect of the interaction between milk-feeding practices and the HLA-DQ genotype on peripheral lymphocyte subsets and their activation markers in infants at familial risk for CD. METHODS: 170 newborns were classified in 3 different genetic risk groups (high risk, HR; intermediate risk, IR; and low risk, LR) after DQB1 and DQA1 typing. Lymphocyte subsets were studied at the age of 4 months by flow cytometry analysis. RESULTS: 79 infants were receiving exclusive breastfeeding (BF) and 91 partial breastfeeding or formula feeding (FF). Regarding genetic risk, 40 infants were classified in HR group, 75 in IR group and 55 in LR group. Two-way ANOVA did not show significant interactions between the type of milk feeding and genetic risk group on the lymphocyte subsets analysed. One-way ANOVA for milk-feeding practice alone showed that the percentage of CD4 + CD25+ cells was significantly higher in BF group than in FF group (BF, 10.92 ± 2.71; FF, 9.94 ± 2.96; p = 0.026), and absolute counts of CD4 + CD38+ cells were significantly higher in FF group than in BF group (FF, 2,881.23 ± 973.48; BF, 2,557.95 ± 977.06; p = 0.038). One-way ANOVA for genetic risk alone showed that absolute counts of NK cells were significantly higher in IR group than HR and LR groups (IR, 539.24 ± 340.63; HR, 405.01 ± 239.53; LR, 419.86 ± 262.85; p = 0.028). CONCLUSION: Lymphocyte subset profiles in the early stages of life could be modulated by milk-feeding practices and genetic risk separately. Breastfeeding might have a positive immunomodulatory effect on lymphocyte subsets in infants at risk of CD.


Asunto(s)
Lactancia Materna , Enfermedad Celíaca/genética , Fórmulas Infantiles , Subgrupos Linfocitarios/inmunología , Análisis de Varianza , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Celíaca/etiología , Enfermedad Celíaca/prevención & control , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/inmunología , Humanos , Lactante , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/metabolismo , Factores de Riesgo
16.
Gut Microbes ; 15(1): 2166780, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36656562

RESUMEN

Visceral hypersensitivity, a fundamental mechanism of chronic visceral pain disorders, can result from both central or peripheral factors, or their combination. As an important regulator of normal gut function, the gut microbiota has been implicated as a key peripheral factor in the pathophysiology of visceral hypersensitivity. Patients with chronic gastrointestinal disorders, such as irritable bowel syndrome, often present with abdominal pain secondary to adverse reactions to dietary components. As both long- and short-term diets are major determinants of gut microbiota configuration that can result in changes in microbial metabolic output, it is becoming increasingly recognized that diet-microbiota interactions play an important role in the genesis of visceral sensitivity. Changes in pain signaling may occur via diet-induced changes in secretion of mediators by both the microbiota and/or host cells. This review will examine the peripheral influence of diet-microbiota interactions underlying increased visceral sensitivity.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Microbiota , Humanos , Microbioma Gastrointestinal/fisiología , Dieta , Percepción
17.
Gut Microbes ; 15(1): 2233679, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37464450

RESUMEN

Compositional changes in the microbiota (dysbiosis) may be a basis for Irritable Bowel Syndrome (IBS), but biomarkers are currently unavailable to direct microbiota-directed therapy. We therefore examined whether changes in fecal ß-defensin could be a marker of dysbiosis in a murine model. Experimental dysbiosis was induced using four interventions relevant to IBS: a mix of antimicrobials, westernized diets (high-fat/high-sugar and high salt diets), or mild restraint stress. Fecal mouse ß-defensin-3 and 16S rRNA-based microbiome profiles were assessed at baseline and during and following these interventions. Each intervention, except for mild restraint stress, altered compositional and diversity profiles of the microbiota. Exposure to antimicrobials or a high-fat/high-sugar diet, but not mild restraint stress, resulted in decreased fecal ß-defensin-3 compared to baseline. In contrast, exposure to the high salt diet increased ß-defensin-3 compared to baseline. Mice exposed to the mix of antimicrobials showed the largest compositional changes and the most significant correlations between ß-defensin-3 levels and bacterial diversity. The high salt diet was also associated with significant correlations between changes in ß-defensin-3 and bacterial diversity, and this was not accompanied by discernible inflammatory changes in the host. Thus, dietary change or antimicrobial exposure, both recognized factors in IBS exacerbations, induced marked dysbiosis that was accompanied by changes in fecal ß-defensin-3 levels. We propose that serial monitoring of fecal ß-defensins may serve as a marker of dysbiosis and help identify those IBS patients who may benefit from microbiota-directed therapeutic interventions.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , beta-Defensinas , Animales , Humanos , Ratones , Dieta Alta en Grasa , Disbiosis/microbiología , Heces/microbiología , Síndrome del Colon Irritable/microbiología , ARN Ribosómico 16S/genética , Azúcares
18.
Gut Microbes ; 15(1): 2188874, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36939195

RESUMEN

Abdominal pain is common in patients with gastrointestinal disorders, but its pathophysiology is unclear, in part due to poor understanding of basic mechanisms underlying visceral sensitivity. Accumulating evidence suggests that gut microbiota is an important determinant of visceral sensitivity. Clinical and basic research studies also show that sex plays a role in pain perception, although the precise pathways are not elucidated. We investigated pain responses in germ-free and conventionally raised mice of both sexes, and assessed visceral sensitivity to colorectal distension, neuronal excitability of dorsal root ganglia (DRG) neurons and the production of substance P and calcitonin gene-related peptide (CGRP) in response to capsaicin or a mixture of G-protein coupled receptor (GPCR) agonists. Germ-free mice displayed greater in vivo responses to colonic distention than conventional mice, with no differences between males and females. Pretreatment with intracolonic capsaicin or GPCR agonists increased responses in conventional, but not in germ-free mice. In DRG neurons, gut microbiota and sex had no effect on neuronal activation by capsaicin or GPCR agonists. While stimulated production of substance P by DRG neurons was similar in germ-free and conventional mice, with no additional effect of sex, the CGRP production was higher in germ-free mice, mainly in females. Absence of gut microbiota increases visceral sensitivity to colorectal distention in both male and female mice. This is, at least in part, due to increased production of CGRP by DRG neurons, which is mainly evident in female mice. However, central mechanisms are also likely involved in this process.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Animales , Femenino , Masculino , Ratones , Péptido Relacionado con Gen de Calcitonina/análisis , Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/farmacología , Sustancia P/análisis , Sustancia P/metabolismo
19.
Gastroenterology ; 141(4): 1314-22, 1322.e1-5, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21745447

RESUMEN

BACKGROUND & AIMS: Proton pump inhibitors (PPIs) and nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used classes of drugs, with the former frequently coprescribed to reduce gastroduodenal injury caused by the latter. However, suppression of gastric acid secretion by PPIs is unlikely to provide any protection against the damage caused by NSAIDs in the more distal small intestine. METHODS: Rats were treated with antisecretory doses of omeprazole or lanzoprazole for 9 days, with concomitant treatment with anti-inflammatory doses of naproxen or celecoxib on the final 4 days. Small intestinal damage was blindly scored, and changes in hematocrit were measured. Changes in small intestinal microflora were evaluated by denaturing gradient gel electrophoresis and reverse-transcription polymerase chain reaction. RESULTS: Both PPIs significantly exacerbated naproxen- and celecoxib-induced intestinal ulceration and bleeding in the rat. Omeprazole treatment did not result in mucosal injury or inflammation; however, there were marked shifts in numbers and types of enteric bacteria, including a significant reduction (∼80%) of jejunal Actinobacteria and Bifidobacteria spp. Restoration of small intestinal Actinobacteria numbers through administration of selected (Bifidobacteria enriched) commensal bacteria during treatment with omeprazole and naproxen prevented intestinal ulceration/bleeding. Colonization of germ-free mice with jejunal bacteria from PPI-treated rats increased the severity of NSAID-induced intestinal injury, as compared with mice colonized with bacteria from vehicle-treated rats. CONCLUSIONS: PPIs exacerbate NSAID-induced intestinal damage at least in part because of significant shifts in enteric microbial populations. Prevention or reversal of this dysbiosis may be a viable option for reducing the incidence and severity of NSAID enteropathy.


Asunto(s)
Actinobacteria/efectos de los fármacos , Antiinflamatorios no Esteroideos/toxicidad , Bifidobacterium/efectos de los fármacos , Hemorragia Gastrointestinal/inducido químicamente , Yeyuno/efectos de los fármacos , Úlcera Péptica/inducido químicamente , Inhibidores de la Bomba de Protones/toxicidad , 2-Piridinilmetilsulfinilbencimidazoles/toxicidad , Actinobacteria/genética , Actinobacteria/crecimiento & desarrollo , Actinobacteria/aislamiento & purificación , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/aislamiento & purificación , Celecoxib , Colon/efectos de los fármacos , Colon/microbiología , Electroforesis en Gel de Gradiente Desnaturalizante , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Hemorragia Gastrointestinal/microbiología , Hemorragia Gastrointestinal/patología , Hemorragia Gastrointestinal/prevención & control , Hematócrito , Yeyuno/microbiología , Yeyuno/patología , Lansoprazol , Masculino , Naproxeno/toxicidad , Omeprazol/toxicidad , Úlcera Péptica/microbiología , Úlcera Péptica/patología , Úlcera Péptica/prevención & control , Probióticos/farmacología , Pirazoles/toxicidad , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/toxicidad , Factores de Tiempo
20.
Clin Dev Immunol ; 2012: 654143, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23008734

RESUMEN

Celiac disease (CD) is an immune-mediated enteropathy, triggered by dietary wheat gluten and similar proteins of barley and rye in genetically susceptible individuals. The etiology of this disorder is complex, involving both environmental and genetic factors. The major genetic risk factor for CD is represented by HLA-DQ genes, which account for approximately 40% of the genetic risk; however, only a small percentage of carriers develop the disease. Gluten is the main environmental factor responsible for the signs and symptoms of the disease, but exposure to gluten does not fully explain the manifestation of CD. Epidemiological and clinical data suggest that environmental factors other than gluten might play a role in disease development, including early feeding practices (e.g., breast milk versus formula and duration of breastfeeding), infections, and alterations in the intestinal microbiota composition. Herein, we review what is known about the influence of dietary factors, exposure to infectious agents, and intestinal microbiota composition, particularly in early life, on the risk of developing CD, as well as the possible dietary strategies to induce or increase gluten tolerance.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Dieta , Intestinos/inmunología , Intestinos/microbiología , Metagenoma/inmunología , Animales , Conducta Alimentaria , Humanos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA