Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Nutr ; 148(6): 910-916, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29767743

RESUMEN

Background: Metabolic endotoxemia is associated with obesity and contributes to postprandial inflammation. Objective: We aimed to determine if low-fat yogurt consumption prevents postprandial inflammation and dysmetabolism in healthy women by inhibiting biomarkers of metabolic endotoxemia. Methods: Premenopausal women defined as obese and nonobese [body mass index (BMI, in kg/m2) 30-40 and 18.5-27, respectively, n = 120] were randomly assigned to consume 339 g of low-fat yogurt (YN, yogurt nonobese; YO, yogurt obese) or 324 g of soy pudding (CN, control nonobese; CO, control obese) for 9 wk (n = 30/group). The intervention foods each supplied 330 kcal with 3 g fat, 66 g carbohydrate, and 4-6 g protein. At weeks 0 and 9, participants ingested 226 g of yogurt or 216 g of soy pudding before a meal providing 56-60 g fat, 82 g carbohydrate, and 28-30 g protein. Plasma soluble CD14 (sCD14), lipopolysaccharide-binding protein (LBP), LPS activity, interleukin-6 (IL-6), glucose, triglyceride, and insulin were measured hourly for 4 h to assess differences in postprandial responses between groups by 2-factor ANOVA. Results: Premeal yogurt consumption prevented the postprandial decrease in sCD14 net incremental area under the curve (net iAUC) by 72% in obese individuals at week 0 (P = 0.0323). YN and YO had ≥40% lower net iAUC of LBP-to-sCD14 ratio and plasma IL-6 concentration than CN and CO, respectively (P < 0.05). CO had postprandial hyperglycemia which was not evident in YO; in contrast YN had 57% less postprandial hypoglycemia than did CN (P-interaction = 0.0013). After 9 wk of yogurt consumption, ΔAUC of LBP-to-sCD14 ratios of YO and YN were less than half of those of the control groups (P = 0.0093). Conclusion: Yogurt consumption improved postprandial metabolism and biomarkers of metabolic endotoxemia in healthy premenopausal women. Premeal yogurt consumption is a feasible strategy to inhibit postprandial dysmetabolism and thus may reduce cardiometabolic risk. This trial was registered at clinicaltrials.gov as NCT01686204.


Asunto(s)
Endotoxinas/toxicidad , Inflamación/sangre , Comidas , Premenopausia , Yogur , Biomarcadores/sangre , Femenino , Humanos , Interleucina-6 , Obesidad , Periodo Posprandial
2.
Crit Rev Food Sci Nutr ; 57(8): 1569-1583, 2017 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-25875150

RESUMEN

Obesity is associated with increased risk for chronic diseases, and affects both developed and developing nations. Yogurt is a nutrient-dense food that may benefit individuals with lactose intolerance, constipation and diarrheal diseases, hypertension, cardiovascular diseases, diabetes, and certain types of cancer. Emerging evidence suggests that yogurt consumption might also improve the health of obese individuals. Obesity is often accompanied by chronic, low-grade inflammation perpetuated by adipose tissue and the gut. In the gut, obesity-associated dysregulation of microbiota and impaired gut barrier function may increase endotoxin exposure. Intestinal barrier function can be compromised by pathogens, inflammatory cytokines, endocannabinoids, diet, exercise, and gastrointestinal peptides. Yogurt consumption may improve gut health and reduce chronic inflammation by enhancing innate and adaptive immune responses, intestinal barrier function, lipid profiles, and by regulating appetite. While this evidence suggests that yogurt consumption is beneficial for obese individuals, randomized-controlled trials are needed to further support this hypothesis.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Obesidad/epidemiología , Yogur/microbiología , Tejido Adiposo/metabolismo , Animales , Apetito , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Enfermedad Crónica , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/prevención & control , Dieta , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina A/metabolismo , Inflamación/etiología , Inflamación/prevención & control , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Metaanálisis como Asunto , Neoplasias/etiología , Neoplasias/prevención & control , Valor Nutritivo , Obesidad/complicaciones , Probióticos/análisis , Factores de Riesgo
3.
Br J Nutr ; 118(12): 1043-1051, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29179781

RESUMEN

The anti-inflammatory mechanisms of low-fat dairy product consumption are largely unknown. The objective of this study was to determine whether low-fat yogurt reduces biomarkers of chronic inflammation and endotoxin exposure in women. Premenopausal women (BMI 18·5-27 and 30-40 kg/m2) were randomised to consume 339 g of low-fat yogurt (yogurt non-obese (YN); yogurt obese (YO)) or 324 g of soya pudding (control non-obese; control obese (CO)) daily for 9 weeks (n 30/group). Fasting blood samples were analysed for IL-6, TNF-α/soluble TNF II (sTNF-RII), high-sensitivity C-reactive protein, 2-arachidonoyl glycerol, anandamide, monocyte gene expression, soluble CD14 (sCD14), lipopolysaccharide (LPS), LPS binding protein (LBP), IgM endotoxin-core antibody (IgM EndoCAb), and zonulin. BMI, waist circumference and blood pressure were also determined. After 9-week yogurt consumption, YO and YN had decreased TNF-α/sTNFR-RII. Yogurt consumption increased plasma IgM EndoCAb regardless of obesity status. sCD14 was not affected by diet, but LBP/sCD14 was lowered by yogurt consumption in both YN and YO. Yogurt intervention increased plasma 2-arachidonoylglycerol in YO but not YN. YO peripheral blood mononuclear cells expression of NF-κB inhibitor α and transforming growth factor ß1 increased relative to CO at 9 weeks. Other biomarkers were unchanged by diet. CO and YO gained approximately 0·9 kg in body weight. YO had 3·6 % lower diastolic blood pressure at week 3. Low-fat yogurt for 9 weeks reduced biomarkers of chronic inflammation and endotoxin exposure in premenopausal women compared with a non-dairy control food. This trial was registered as NCT01686204.


Asunto(s)
Biomarcadores/sangre , Dieta , Endotoxinas/toxicidad , Inflamación/sangre , Inflamación/dietoterapia , Yogur/análisis , Proteínas de Fase Aguda , Adulto , Antropometría , Ácidos Araquidónicos/sangre , Proteína C-Reactiva/metabolismo , Proteínas Portadoras/sangre , Enfermedad Crónica , Citocinas/sangre , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/análisis , Endocannabinoides/sangre , Endotoxemia/sangre , Endotoxemia/dietoterapia , Femenino , Glicéridos/sangre , Humanos , Inmunoglobulina M/sangre , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/sangre , Persona de Mediana Edad , FN-kappa B/metabolismo , Obesidad/metabolismo , Alcamidas Poliinsaturadas/sangre , Adulto Joven
4.
Food Chem ; 359: 129831, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-33957324

RESUMEN

The objective of this study was to determine the extent that the aronia berry matrix affects gut microbiota composition, fecal short chain fatty acids (SCFAs), and colonic anthocyanins in healthy mice. C57BL/6J mice were fed AIN-93 M control diet (C) or C with whole aronia berry (AB), aronia extract (AE), or polyphenol-depleted AB (D) at the expense of cornstarch. After one week of feeding, AB and D increased fecal anthocyanins more than AE. Diets differentially affected SCFA and microbiota. AB fecal SCFA was associated with increased metabolism of succinate and pyruvate to butyrate. D increased acetic acid production, was associated with increased abundance of predicted genes for fermentation of carbohydrates to acetyl-coA. AB and D also increased predicted abundance of microbial catechol metabolism pathway I relative to C, which was attributed to enrichment of Lachnospiraceae. Therefore, the berry matrix impacts how aronia polyphenols interact with the gut microbiota in healthy mice.


Asunto(s)
Frutas/química , Microbioma Gastrointestinal/efectos de los fármacos , Photinia/química , Polifenoles/farmacología , Animales , Colon/efectos de los fármacos , Colon/microbiología , Heces/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Mol Nutr Food Res ; 63(5): e1800985, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30521111

RESUMEN

SCOPE: Increased fruit consumption is associated with reduced risk of colitis. It has been investigated whether the anti-colitic effects of the polyphenol-rich aronia berry (Aronia mitschurinii 'Viking') are mediated through Th17 and Treg. METHODS AND RESULTS: Colitis is induced in recombinase activating gene-1 deficient mice injected with syngeneic CD4+ CD62L+ naïve T cells. Mice consume either 4.5% w/w aronia-berry-supplemented or a control diet concurrent with T cell transfer. The extent of colitis and immunocyte populations are evaluated at weeks 3 to 7 after transfer. Aronia consumption prevents colitic wasting and reduces colon weight/length ratios relative to the control diet at weeks 5 and 7. Compared to the control diet, aronia feeding increases Treg in mesenteric lymph node at all colitis stages. Treg and regulatory Th17 subpopulations (IL-17A+ IL-10+ and IL-17A+ IL-22+ ) are increased in lamina propria and spleen at week 5 in aronia-fed mice. Aronia feeding also decreases total CD4+ cells but increases colonic Tregs. The ability of aronia to modulate colonic cytokines is associated with functional T cell IL-10 and increased diversity of microbiota. CONCLUSIONS: Aronia berry consumption inhibits adoptive transfer colitis by increasing Treg and regulatory Th17 cells. Dietary modulation of T cells is dynamic and precedes colitic wasting.


Asunto(s)
Colitis/dietoterapia , Photinia , Linfocitos T Reguladores , Células Th17 , Animales , Linfocitos T CD4-Positivos/patología , Diferenciación Celular , Colitis/patología , Colitis/prevención & control , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Interleucina-10/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Bazo/citología , Linfocitos T Reguladores/metabolismo
6.
Nutrients ; 11(6)2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31212794

RESUMEN

Oxidative stress is involved in the pathogenesis and progression of inflammatory bowel disease. Consumption of aronia berry inhibits T cell transfer colitis, but the antioxidant mechanisms pertinent to immune function are unclear. We hypothesized that aronia berry consumption could inhibit inflammation by modulating the antioxidant function of immunocytes and gastrointestinal tissues. Colitis was induced in recombinase activating gene-1 deficient (Rag1-/-) mice injected with syngeneic CD4+CD62L+ naïve T cells. Concurrent with transfer, mice consumed either 4.5% w/w aronia berry-supplemented or a control diet for five weeks. Aronia berry inhibited intestinal inflammation evidenced by lower colon weight/length ratios, 2-deoxy-2-[18F]fluoro-d-glucose (FDG) uptake, mRNA expressions of tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) in the colon. Aronia berry also suppressed systemic inflammation evidenced by lower FDG uptake in the spleen, liver, and lung. Colitis induced increased colon malondialdehyde (MDA), decreased colon glutathione peroxidase (GPx) activity, reduced glutathione (rGSH) level, and suppressed expression of antioxidant enzymes in the colon and mesenteric lymph node (MLN). Aronia berry upregulated expression of antioxidant enzymes, prevented colitis-associated depletion of rGSH, and maintained GPx activity. Moreover, aronia berry modulated mitochondria-specific antioxidant activity and decreased splenic mitochondrial H2O2 production in colitic mice. Thus, aronia berry consumption inhibits oxidative stress in the colon during T cell transfer colitis because of its multifaceted antioxidant function in both the cytosol and mitochondria of immunocytes.


Asunto(s)
Antioxidantes/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Colitis/inmunología , Suplementos Dietéticos , Frutas , Estrés Oxidativo/efectos de los fármacos , Photinia , Animales , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Inflamación , Interferón gamma/metabolismo , Intestinos/inmunología , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
7.
Sci Transl Med ; 10(443)2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29848663

RESUMEN

Triclosan (TCS) is a high-volume chemical used as an antimicrobial ingredient in more than 2000 consumer products, such as toothpaste, cosmetics, kitchenware, and toys. We report that brief exposure to TCS, at relatively low doses, causes low-grade colonic inflammation, increases colitis, and exacerbates colitis-associated colon cancer in mice. Exposure to TCS alters gut microbiota in mice, and its proinflammatory effect is attenuated in germ-free mice. In addition, TCS treatment increases activation of Toll-like receptor 4 (TLR4) signaling in vivo and fails to promote colitis in Tlr4-/- mice. Together, our results demonstrate that this widely used antimicrobial ingredient could have adverse effects on colonic inflammation and associated colon tumorigenesis through modulation of the gut microbiota and TLR4 signaling. Together, these results highlight the need to reassess the effects of TCS on human health and potentially update policies regulating the use of this widely used antimicrobial.


Asunto(s)
Antiinfecciosos/efectos adversos , Carcinogénesis/patología , Colitis/complicaciones , Colon/patología , Neoplasias del Colon/inducido químicamente , Inflamación/inducido químicamente , Animales , Colitis/microbiología , Colitis/patología , Colon/microbiología , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/microbiología , Inflamación/patología , Masculino , Metaboloma , Ratones Endogámicos C57BL , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Triclosán/efectos adversos
8.
Food Funct ; 6(6): 1773-86, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25986932

RESUMEN

Crohn's disease and ulcerative colitis presently have no cure and are treated with anti-inflammatory drugs or monoclonal antibodies targeting pro-inflammatory cytokines. A variety of rodent models have been used to model chronic and acute colitis. Dietary polyphenols in foods and botanicals are of considerable interest for prevention and treatment of colitis. Many dietary polyphenols have been utilized for prevention of colitis in rodent models. Berries, green tea polyphenols, curcumin, and stilbenes have been the most extensively tested polyphenols in rodent models of colitis. The majority of polyphenols tested have inhibited colitis in rodents, but increasing doses of EGCG and green tea, isoflavones, flaxseed, and α-mangostin have exacerbated colitis. Few studies have examined combination of polyphenols or other bioactives for inhibition of colitis. Translating polyphenol doses used in rodent models of colitis to human equivalent doses reveals that supplemental doses are most likely required to inhibit colitis from a single polyphenol treatment. The ability to translate polyphenol treatments in rodent models is likely to be limited by species differences in xenobiotic metabolism and microbiota. Given these limitations, data from polyphenols in rodent models suggests merit for pursuing additional clinical studies for prevention of colitis.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Flavonoides/uso terapéutico , Alimentos Funcionales , Enfermedades Inflamatorias del Intestino/prevención & control , Polifenoles/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/efectos adversos , Ácidos Cumáricos/administración & dosificación , Ácidos Cumáricos/efectos adversos , Ácidos Cumáricos/uso terapéutico , Suplementos Dietéticos/efectos adversos , Flavonoides/administración & dosificación , Flavonoides/efectos adversos , Fármacos Gastrointestinales/administración & dosificación , Fármacos Gastrointestinales/efectos adversos , Fármacos Gastrointestinales/uso terapéutico , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/dietoterapia , Enfermedades Inflamatorias del Intestino/inmunología , Fenoles/administración & dosificación , Fenoles/efectos adversos , Fenoles/uso terapéutico , Extractos Vegetales/administración & dosificación , Extractos Vegetales/efectos adversos , Extractos Vegetales/uso terapéutico , Polifenoles/administración & dosificación , Polifenoles/efectos adversos , Estilbenos/administración & dosificación , Estilbenos/efectos adversos , Estilbenos/uso terapéutico , Xantonas/administración & dosificación , Xantonas/efectos adversos , Xantonas/uso terapéutico
9.
Nutr Res ; 33(5): 406-13, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23684442

RESUMEN

We hypothesized that a polyphenol-rich chokeberry extract (CBE) would modulate hepatic lipid metabolism and improve antioxidant function in apolipoprotein E knockout (apoE(-/-)) mice. ApoE(-/-) mice were fed diets containing 15% fat with 0.2% cholesterol alone or supplemented with 0.005% or 0.05% CBE for 4 weeks. CBE polyphenol content was determined by the total phenols, 4-dimethylaminocinnamaldehyde, and ultra high-performance liquid chromatography-mass spectrometry methods. The 0.05% CBE diet provided mice with mean daily doses of 1.2 mg gallic acid equivalents of total phenols, 0.19 mg anthocyanins, 0.17 mg phenolic acids, 0.06 mg proanthocyanidins (as catechin-equivalents), and 0.02 mg flavonols. The 0.05% CBE group had 12% less plasma total cholesterol concentrations than the control. Despite the hypocholesterolemic effect of CBE, hepatic mRNA levels of low-density lipoprotein receptor, hydroxyl-3-methylglutaryl coenzyme A reductase and cholesterol 7α-hydroxylase in CBE-fed mice were not significantly different from controls. Dietary CBE did not alter hepatic lipid content or the hepatic expression of genes involved in lipogenesis and fatty acid ß-oxidation such as fatty acid synthase, carnitine palmitoyltransferase 1 and acyl-CoA oxidase. Plasma paraoxonase and catalase activities were significantly increased in mice fed 0.05% CBE. Both CBE diets increased hepatic glutathione peroxidase (GPx) activity but the 0.05% CBE group had 24% less proximal intestine GPx activity relative to controls. Thus, dietary CBE lowered total cholesterol and improved plasma and hepatic antioxidant function at nutritionally-relevant doses in apoE(-/-) mice. Furthermore, the CBE cholesterol-lowering mechanism in apoE(-/-) mice was independent of hepatic expression of genes involved in cholesterol metabolism.


Asunto(s)
Antioxidantes/metabolismo , Apolipoproteínas E/genética , Colesterol/sangre , Photinia/química , Extractos Vegetales/farmacología , Polifenoles/farmacología , Acil-CoA Oxidasa/genética , Acil-CoA Oxidasa/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Cromatografía Líquida de Alta Presión , Cinamatos/farmacología , Dieta Alta en Grasa , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA