Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nutrients ; 13(7)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209042

RESUMEN

BACKGROUND AND AIMS: Altering dietary ferrous sulphate (FS) consumption exacerbates a murine model of colitis and alters the intestinal microbiome. We investigated the impact of oral ferric maltol (FM) and FS on mice with dextran sodium sulphate (DSS) induced colitis, and the microbiome of patients with iron deficiency. METHODS: Mice had acute colitis induced, with 2% DSS for 5 days, followed by water. During this period, groups of mice were fed standard chow (200 ppm iron, SC, n = 8), or SC with 200ppm FS supplementation (n = 16, FSS), or SC with 200 ppm FM supplementation (n = 16, FMS). Clinical, pathological and microbiome assessments were compared at days 1 and 10. Fecal bacterial gDNA was extracted and the microbiome assessed by sequencing. Statistical inferences were made using MacQIIME. Principal Coordinates Analysis were used to visualize beta-diversity cluster analysis. Ten patients with IDA were treated with FS, and six with inactive inflammatory bowel disease received FM, supplements for four weeks: pre- and mid-treatment fecal samples were collected: the microbiome was assessed (see above). RESULTS: In mice, after DSS treatment, there was a decrease in many genera in the SC and FSS groups: Lactobacillales increased in mice that received FMS. In humans, FS treatment led to an increase in five genera, but FM was not associated with any measurable change. The severity of DSS-induced colitis was greater with FSS than FMS. CONCLUSIONS: This study demonstrates differential and unique influences of ferric maltol and ferrous sulphate supplements on intestinal microbiota. These differences might contribute to the different side effects associated with these preparations.


Asunto(s)
Compuestos Férricos/administración & dosificación , Compuestos Férricos/farmacología , Compuestos Ferrosos/farmacología , Pironas/administración & dosificación , Pironas/farmacología , Administración Oral , Animales , Biodiversidad , Peso Corporal/efectos de los fármacos , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Colon/efectos de los fármacos , Colon/microbiología , Colon/patología , Sulfato de Dextran , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Hierro/metabolismo , Ratones Endogámicos C57BL , Filogenia
2.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807459

RESUMEN

BACKGROUND: Oral iron supplementation causes gastrointestinal side effects. Short-term alterations in dietary iron exacerbate inflammation and alter the gut microbiota, in murine models of colitis. Patients typically take supplements for months. We investigated the impact of long-term changes in dietary iron on colitis and the microbiome in mice. METHODS: We fed mice chow containing differing levels of iron, reflecting deficient (100 ppm), normal (200 ppm), and supplemented (400 ppm) intake for up to 9 weeks, both in absence and presence of dextran sodium sulphate (DSS)-induced chronic colitis. We also induced acute colitis in mice taking these diets for 8 weeks. Impact was assessed (i) clinically and histologically, and (ii) by sequencing the V4 region of 16S rRNA. RESULTS: In mice with long-term changes, the iron-deficient diet was associated with greater weight loss and histological inflammation in the acute colitis model. Chronic colitis was not influenced by altering dietary iron however there was a change in the microbiome in DSS-treated mice consuming 100 ppm and 400 ppm iron diets, and control mice consuming the 400 ppm iron diet. Proteobacteria levels increased significantly, and Bacteroidetes levels decreased, in the 400 ppm iron DSS group at day-63 compared to baseline. CONCLUSIONS: Long-term dietary iron alterations affect gut microbiota signatures but do not exacerbate chronic colitis, however acute colitis is exacerbated by such dietary changes. More work is needed to understand the impact of iron supplementation on IBD. The change in the microbiome, in patients with colitis, may arise from the increased luminal iron and not simply from colitis.


Asunto(s)
Colitis/metabolismo , Sobrecarga de Hierro/fisiopatología , Hierro/metabolismo , Anemia Ferropénica , Animales , Bacterias/genética , Colitis/fisiopatología , Colon/patología , Sulfato de Dextran/farmacología , Dieta , Suplementos Dietéticos/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/etiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Inflamación , Enfermedades Inflamatorias del Intestino/patología , Hierro de la Dieta/efectos adversos , Ratones , Ratones Endogámicos C57BL , Microbiota , ARN Ribosómico 16S/genética
3.
FASEB J ; 33(3): 3129-3136, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30359099

RESUMEN

Metabolomics studies have the potential to discover biomarkers. Fecal volatile organic compounds (VOCs) have been found to differ in patients with inflammatory bowel disease and irritable bowel syndrome. Murine models of colitis offer an alternative to human studies in which diet can be controlled. We aimed to investigate fecal VOCs from mice in which acute and chronic colitis was induced. Groups of adult C57BL/6 mice underwent treatment with oral dextran sulfate sodium to induce colitis. Control mice received no treatment or had acute osmotic diarrhea induced with magnesium sulfate. Colitis was assessed clinically and by histology. Samples of feces and/or colon contents were collected and volatile compounds determined by solid phase microextraction-GC-MS. Statistics were performed using metabolomics tools. Acute colitis was associated with an increase in aldehydes and chronic colitis with one specific ketone. Osmotic diarrhea was associated with a significant reduction in VOCs, especially alcohols. We provide evidence that the identification of disease-associated VOC concentration ranges, combined with specific marker compounds, would potentially increase the likelihood of finding an inflammatory bowel disease-specific fecal VOC marker profile.-Reade, S., Williams, J. M., Aggio, R., Duckworth, C. A., Mahalhal, A., Hough, R., Pritchard, D. M., Probert, C. S., Potential role of fecal volatile organic compounds as biomarkers of chemically induced intestinal inflammation in mice.


Asunto(s)
Colitis/metabolismo , Heces/química , Compuestos Orgánicos Volátiles/análisis , Enfermedad Aguda , Aldehídos/análisis , Animales , Biomarcadores/análisis , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Diarrea/inducido químicamente , Diarrea/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/metabolismo , Metabolómica , Ratones , Ratones Endogámicos C57BL
4.
PLoS One ; 13(10): e0202460, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30308045

RESUMEN

Inflammatory bowel disease (IBD) is associated with anaemia and oral iron replacement to correct this can be problematic, intensifying inflammation and tissue damage. The intestinal microbiota also plays a key role in the pathogenesis of IBD, and iron supplementation likely influences gut bacterial diversity in patients with IBD. Here, we assessed the impact of dietary iron, using chow diets containing either 100, 200 or 400 ppm, fed ad libitum to adult female C57BL/6 mice in the presence or absence of colitis induced using dextran sulfate sodium (DSS), on (i) clinical and histological severity of acute DSS-induced colitis, and (ii) faecal microbial diversity, as assessed by sequencing the V4 region of 16S rRNA. Increasing or decreasing dietary iron concentration from the standard 200 ppm exacerbated both clinical and histological severity of DSS-induced colitis. DSS-treated mice provided only half the standard levels of iron ad libitum (i.e. chow containing 100 ppm iron) lost more body weight than those receiving double the amount of standard iron (i.e. 400 ppm); p<0.01. Faecal calprotectin levels were significantly increased in the presence of colitis in those consuming 100 ppm iron at day 8 (5.94-fold) versus day-10 group (4.14-fold) (p<0.05), and for the 400 ppm day-8 group (8.17-fold) versus day-10 group (4.44-fold) (p<0.001). In the presence of colitis, dietary iron at 400 ppm resulted in a significant reduction in faecal abundance of Firmicutes and Bacteroidetes, and increase of Proteobacteria, changes which were not observed with lower dietary intake of iron at 100 ppm. Overall, altering dietary iron intake exacerbated DSS-induced colitis; increasing the iron content of the diet also led to changes in intestinal bacteria diversity and composition after colitis was induced with DSS.


Asunto(s)
Anemia/tratamiento farmacológico , Colitis/dietoterapia , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Hierro de la Dieta/administración & dosificación , Hierro/metabolismo , Administración Oral , Anemia/microbiología , Anemia/patología , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Ratones , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...