Mostrar el registro sencillo del ítem

dc.contributor.advisorRey, Federico
dc.contributor.advisorAcosta González, Luis Alejandro
dc.contributor.authorRodríguez Castano, Gina Paola del Carmen
dc.date.accessioned5/14/2020 11:40
dc.date.available5/14/2020 11:40
dc.date.issued2020-03-13
dc.identifier.urihttp://hdl.handle.net/10818/41023
dc.description135 páginases_CO
dc.description.abstractFlavonoids are secondary metabolites of plants which have anti-cancer, anti-inflammatory, and antioxidant properties. However, the intestinal microbiota can change the bioactivity and bioavailability of these compounds, which may trigger different levels of response to a treatment. In order to expand our understanding of the capacity of the gut microbiota to modify these therapeutic compounds, we explored the microbial degradation of quercetin, one the most abundant flavonoids in the human diet. First, we revealed that a non-quercetin degrader (Bacteroides thetaiotaomicron) can provide, via crossfeeding, substrates to a quercetin-degrader (Eubacterium ramulus) for the cometabolization of the flavonoid. Second, through a metataxonomic analysis of fecal communities exposed to the flavonoid, we detected two variants related to the quercetin degrader, Flavonifractor plautii, that presented a negative correlation in their relative abundances upon incubation with quercetin. Lastly, a bioinformatic analysis of the genome of the closest relatives of these variantsshowed that they are discordant for the catabolism of an important substrate in the gastrointestinal tract, ethanolamine, which it is formed from bacterial and intestinal cell membranes and is abundant even in the absence of dietary compounds due to the constant washing away of these cells in the intestinal mucus. Overall, these observations indicate that flavonoid-degrading bacteria can be differentially affected by dietary and host’s substrates and interactions with different microbial species. Thus, the community structure and metabolic capacity of each individual’s gut microbiota may impact the health-related effects of these compounds.en
dc.formatapplication/pdfes_CO
dc.language.isoenges_CO
dc.publisherUniversidad de La Sabanaes_CO
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 International*
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/*
dc.sourceUniversidad de La Sabana
dc.sourceIntellectum Repositorio Universidad de La Sabana
dc.subjectFlavonoideses_CO
dc.subjectMetabolitos microbianoses_CO
dc.subjectMicrobioma gastrointestinales_CO
dc.subjectMembranas plasmáticases_CO
dc.titleStudy on the interaction between diet, quercetin and intestinal microbiotaen
dc.typedoctoral thesises_CO
dc.identifier.local276807
dc.identifier.localTE10643
dc.type.hasVersionpublishedVersiones_CO
dc.rights.accessRightsopenAccesses_CO
dcterms.referencesA. Drewnowski and B. M. Popkin, “The Nutrition Transition: New Trends in the Global Diet,” Nutr. Rev., vol. 55, no. 2, pp. 31–43, 1997.en
dcterms.referencesF. B. Hu et al., “Reproducibility and validity of dietary patterns assessed with a food-frequency questionnaire,” Am. J. Clin. Nutr., vol. 69, no. 2, pp. 243–249, 1999.en
dcterms.referencesM. L. Slattery, K. M. Boucher, B. J. Caan, J. D. Potter, and K. N. Ma, “Eating patterns and risk of colon cancer.,” Am. J. Epidemiol., vol. 148, no. 1, pp. 4–16, 1998.en
dcterms.referencesA. Nanri et al., “Reproducibility and validity of dietary patterns assessed by a food frequency questionnaire used in the 5-year follow-up survey of the Japan Public Health Center-Based Prospective Study,” J. Epidemiol., vol. 22, no. 3, pp. 205–215, 2012en
dcterms.referencesL. Cordain et al., “Origins and evolution of the Western diet: health implications for the 21st century.,” Am. J. Clin. Nutr., vol. 81, no. 2, pp. 341–54, 2005.en
dcterms.referencesW. Price, “Nutrition and Physical Degeneration,” Can. Med. Assoc. J., vol. 42, no. 2, p. 208, 1939.en
dcterms.referencesS. Parry and L. Straker, “The contribution of office work to sedentary behaviour associated risk,” BMC Public Health, vol. 13, no. 1, pp. 1–10, 2013.en
dcterms.referencesCDC, “Vital signs: binge drinking prevalence, frequency, and intensity among adults - United States, 2010.,” 2012.en
dcterms.referencesWHO, “Global status report on noncommunicable diseases 2014,” World Health, p. 176, 2014.en
dcterms.referencesS. Kanoski and T. Davidson, “Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity,” Physiol. Behav., vol. 103, no. 1, pp. 59–68, 2011.en
dcterms.referencesL. Bazzano, “Dietary intake of fruit and vegetables and risk of diabetes mellitus and cardiovascular diseases,” Geneva WHO, 2005.en
dcterms.referencesF. M. Trovato, D. Catalano, G. F. Martines, P. Pace, and G. M. Trovato, “Mediterranean diet and non-alcoholic fatty liver disease. The need of extended and comprehensive interventions,” Clin. Nutr., vol. 34, no. 1, pp. 86–88, 2015.en
dcterms.referencesJ. M. Geleijnse, F. J. Kok, and D. E. Grobbee, “Impact of dietary and lifestyle factors on the prevalence of hypertension in Western populations,” Eur. J. Public Health, vol. 14, no. 3, pp. 235–239, 2004.en
dcterms.referencesS. Jehle, A. Zanetti, J. Muser, H. N. Hulter, and R. Krapf, “Partial neutralization of the acidogenic Western diet with potassium citrate increases bone mass in postmenopausal women with osteopenia.,” J. Am. Soc. Nephrol., vol. 17, no. 11, pp. 3213–3222, 2006.en
dcterms.referencesA. Manzel, D. N. Muller, D. A. Hafler, S. E. Erdman, R. A. Linker, and M. Kleinewietfeld, “Role of ‘western diet’ in inflammatory autoimmune diseases,” Curr. Allergy Asthma Rep., vol. 14, no. 1, pp. 1–8, 2014.en
dcterms.referencesH. Adlercreutz, “Western diet and Western diseases: Some hormonal and biochemical mechanisms and associations,” Scand. J. Clin. Lab. Invest. Suppl., vol. 50, no. sup201, pp. 3–23, 1990.en
dcterms.referencesJ. Lederberg and A. McCray, “Ome SweetOmics--A Genealogical Treasury of Words,” Sci., vol. 15, no. 7, p. 8, 2001.en
dcterms.referencesC. Huttenhower et al., “Structure, function and diversity of the healthy human microbiome,” Nature, vol. 486, no. 7402, pp. 207–214, 2012.en
dcterms.referencesP. B. Eckburg et al., “Diversity of the human intestinal microbial flora.,” Science, vol. 308, no. 5728, pp. 1635– 8, 2005en
dcterms.referencesJ. Qin, R. Li, J. Raes, and M. Arumugam, “A human gut microbial gene catalogue established by metagenomic sequencing,” Nature, vol. 464, no. 7285, pp. 59–65, 2010.en
dcterms.referencesM. Frémont, D. Coomans, S. Massart, and K. De Meirleir, “High-throughput 16S rRNA gene sequencing reveals alterations of intestinal microbiota in myalgic encephalomyelitis/chronic fatigue syndrome patients,” Anaerobe, vol. 22, pp. 50–56, 2013.en
dcterms.referencesX. Zhang et al., “The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment.,” Nat. Med., vol. 21, no. 8, pp. 895–905, 2015.en
dcterms.referencesC. T. Brown et al., “Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes,” PLoS One, vol. 6, no. 10, 2011.en
dcterms.referencesN. Larsen et al., “Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults,” PLoS One, vol. 5, no. 2, 2010.en
dcterms.referencesK. Forslund et al., “Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota.,” Nature, vol. 528, no. 7581, pp. 262–266, 2015.en
dcterms.referencesM. Joossens et al., “Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives,” Gut, vol. 60, no. 5, pp. 631–637, 2011.en
dcterms.referencesA. R. Erickson et al., “Integrated Metagenomics/Metaproteomics Reveals Human Host-Microbiota Signatures of Crohn’s Disease,” PLoS One, vol. 7, no. 11, 2012.en
dcterms.referencesI. A. Finnie, A. D. Dwarakanath, B. A. Taylor, and J. M. Rhodes, “Colonic mucin synthesis is increased by sodium butyrate.,” Gut, vol. 36, no. 1, pp. 93–9, 1995.en
dcterms.referencesL. Peng, Z.-R. Li, R. S. Green, I. R. Holzman, and J. Lin, “Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers.,” J. Nutr., vol. 139, no. 9, pp. 1619–1625, 2009.en
dcterms.referencesM. A. R. Vinolo, H. G. Rodrigues, R. T. Nachbar, and R. Curi, “Regulation of Inflammation by Short Chain Fatty Acids,” Nutrients, vol. 3, no. 12, pp. 858–876, 2011.en
dcterms.referencesP. Louis, P. Young, G. Holtrop, and H. J. Flint, “Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene,” Environ. Microbiol., vol. 12, no. 2, pp. 304–314, 2010.en
dcterms.referencesM. J. Hopkins, R. Sharp, and G. T. Macfarlane, “Age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles.,” Gut, vol. 48, no. 2, pp. 198–205, 2001.en
dcterms.referencesP. J. Turnbaugh et al., “A core gut microbiome in obese and lean twins.,” Nature, vol. 457, no. 7228, pp. 480– 484, 2009.en
dcterms.referencesL. C. Kong et al., “Gut microbiota after gastric bypass in human obesity: Increased richness and associations of bacterial genera with adipose tissue genes,” Am. J. Clin. Nutr., vol. 98, no. 1, pp. 16–24, 2013.en
dcterms.referencesC. de Weerth, S. Fuentes, P. Puylaert, and W. M. de Vos, “Intestinal microbiota of infants with colic: development and specific signatures.,” Pediatrics, vol. 131, no. 2, pp. e550-8, 2013.en
dcterms.references] T. Yatsunenko et al., “Human gut microbiome viewed across age and geography,” Nature, vol. 486, no. 7402, pp. 222–227, 2012.en
dcterms.referencesC. A. Lozupone, M. E. Rhodes, C. P. Neff, A. P. Fontenot, T. B. Campbell, and B. E. Palmer, “HIV-induced alteration in gut microbiota,” Gut Microbes, vol. 5, no. 4, pp. 562–570, 2014en
dcterms.referencesW. H. W. Tang et al., “Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk.,” N. Engl. J. Med., vol. 368, no. 17, pp. 1575–84, 2013.en
dcterms.referencesR. a Koeth et al., “Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis,” Nat. Med., vol. 19, no. April, pp. 576–85, 2013.en
dcterms.referencesT. Poutahidis et al., “Microbial Reprogramming Inhibits Western Diet-Associated Obesity,” PLoS One, vol. 8, no. 7, p. e68596, 2013.en
dcterms.referencesF. Armougom, M. Henry, B. Vialettes, D. Raccah, and D. Raoult, “Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients,” PLoS One, vol. 4, no. 9, p. e7125, 2009.en
dcterms.referencesM. Million et al., “Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii.,” Int. J. Obes. (Lond)., vol. 36, no. 6, pp. 817–25, 2012.en
dcterms.referencesF. Fåk and F. Bäckhed, “Lactobacillus reuteri prevents diet-induced obesity, but not atherosclerosis, in a strain dependent fashion in Apoe−/− mice,” PLoS One, vol. 7, no. 10, p. e46837, 2012.en
dcterms.referencesM. P. Diaz-Ropero et al., “Two Lactobacillus strains, isolated from breast milk, differently modulate the immune response,” J. Appl. Microbiol., vol. 102, no. 2, pp. 337–343, 2007.en
dcterms.referencesB. T. Burton and W. R. Foster, “Health implications of obesity: an NIH Consensus Development Conference,” J. Am. Diet. Assoc., vol. 85, no. 9, pp. 1117–1121, 1985en
dcterms.referencesC. Ogden, S. Yanovski, M. Carroll, and K. Flegal, “The epidemiology of obesity,” Gastroenterology, vol. 132, no. 6, pp. 2087–2102, 2007.en
dcterms.referencesJ. V Van Vliet-Ostaptchouk et al., “The prevalence of Metabolic Syndrome and metabolically healthy obesity in Europe: a collaborative analysis of ten large cohort studies.,” BMC Endocr. Disord., vol. 14, no. 9, pp. 1– 13, 2014.en
dcterms.referencesN. Klöting et al., “Insulin-sensitive obesity,” Am. J. Physiol. - Endocrinol. Metab., vol. 299, no. 3, pp. 506–515, 2010.en
dcterms.referencesJ. Naukkarinen et al., “Characterising metabolically healthy obesity in weight-discordant monozygotic twins,” Diabetologia, vol. 57, no. 1, pp. 167–176, 2014.en
dcterms.referencesK. Azuma et al., “Higher liver fat content among Japanese in Japan compared with non-Hispanic whites in the United States,” Metabolism., vol. 58, no. 8, pp. 1200–1207, 2009.en
dcterms.referencesR. W. Walker et al., “High rates of fructose malabsorption are associated with reduced liver fat in obese African Americans.,” J. Am. Coll. Nutr., vol. 31, no. 5, pp. 369–74, 2012en
dcterms.referencesR. Guerrero, G. L. Vega, S. M. Grundy, and J. D. Browning, “Ethnic differences in hepatic steatosis: An insulin resistance paradox?,” Hepatology, vol. 49, no. 3, pp. 791–801, 2009.en
dcterms.referencesM. G. Marmot, S. L. Syme, A. Kagan, H. Kato, J. B. Cohen, and J. Belsky, “Epidemiologic studies of coronary heart disease and stroke in Japanese men living in Japan, Hawaii and California: prevalence of coronary and hypertensive heart disease and associated risk factors.,” Am. J. Epidemiol., vol. 102, no. 6, pp. 514–25, 1975.en
dcterms.referencesC. B. Newgard, “Interplay between lipids and branched-chain amino acids in development of insulin resistance,” Cell Metabolism, vol. 15, no. 5. pp. 606–614, 2012.en
dcterms.referencesM. Raman et al., “Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease,” Clin. Gastroenterol. Hepatol., vol. 11, no. 7, pp. 868–875, 2013.en
dcterms.referencesL. Zhu et al., “Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH,” Hepatology, vol. 57, no. 2, pp. 601–609, 2013.en
dcterms.referencesJ. Boursier and A. M. Diehl, “Nonalcoholic Fatty Liver Disease and the Gut Microbiome,” Clinics in Liver Disease, vol. 20, no. 2. pp. 263–275, 2016.en
dcterms.referencesF. Bäckhed, J. K. Manchester, C. F. Semenkovich, and J. I. Gordon, “Mechanisms underlying the resistance to diet-induced obesity in germ-free mice,” Proc. Natl. Acad. Sci., vol. 104, no. 3, pp. 979–984, 2007.en
dcterms.referencesA. M. W. Petersen and B. K. Pedersen, “The anti-inflammatory effect of exercise.,” J. Appl. Physiol., vol. 98, no. 4, pp. 1154–1162, 2005.en
dcterms.referencesJ. M. Wells, O. Rossi, M. Meijerink, and P. van Baarlen, “Epithelial crosstalk at the microbiota-mucosal interface.,” Proc. Natl. Acad. Sci. U. S. A., vol. 108 Suppl, pp. 4607–4614, 2011.en
dcterms.referencesG. G. Muccioli et al., “The endocannabinoid system links gut microbiota to adipogenesis.,” Mol. Syst. Biol., vol. 6, no. 392, p. 392, 2010.en
dcterms.referencesH. Nakarai et al., “Adipocyte-macrophage interaction may mediate LPS-induced low-grade inflammation: potential link with metabolic complications.,” Innate Immun., vol. 18, no. 1, pp. 164–70, 2012.en
dcterms.referencesF. S. Lira et al., “Endotoxin levels correlate positively with a sedentary lifestyle and negatively with highly trained subjects.,” Lipids Health Dis., vol. 9, no. 1, p. 82, 2010.en
dcterms.referencesE. Pace et al., “Cigarette smoke increases Toll-like receptor 4 and modifies lipopolysaccharide-mediated responses in airway epithelial cells,” Immunology, vol. 124, no. 3, pp. 401–411, 2008en
dcterms.referencesM. Lyte and S. Ernst, “Catecholamine induced growth of gram negative bacteria,” Life Sci., vol. 50, no. 3, pp. 203–212, 1992.en
dcterms.referencesP. P. E. Freestone, S. M. Sandrini, R. D. Haigh, and M. Lyte, “Microbial endocrinology: how stress influences susceptibility to infection,” Trends in Microbiology, vol. 16, no. 2. pp. 55–64, 2008en
dcterms.referencesM. Lyte and M. T. Bailey, “Neuroendocrine-bacterial interactions in a neurotoxin-induced model of trauma.,” J. Surg. Res., vol. 70, no. 2, pp. 195–201, 1997.en
dcterms.referencesM. Lyte, C. D. Frank, and B. T. Green, “Production of an autoinducer of growth by norepinephrine cultured Escherichia coli O157:H7,” FEMS Microbiol. Lett., vol. 139, no. 2–3, pp. 155–159, 1996.en
dcterms.referencesP. D. Cani et al., “Metabolic endotoxemia initiates obesity and insulin resistance,” Diabetes, vol. 56, no. 7, pp. 1761–1772, 2007.en
dcterms.referencesS. Ghoshal, J. Witta, J. Zhong, W. de Villiers, and E. Eckhardt, “Chylomicrons promote intestinal absorption of lipopolysaccharides.,” J. Lipid Res., vol. 50, no. 1, pp. 90–97, 2009.en
dcterms.referencesS. Sadeghi, F. A. Wallace, and P. C. Calder, “Dietary lipids modify the cytokine response to bacterial lipopolysaccharide in mice,” Immunology, vol. 96, no. 3, pp. 404–410, 1999.en
dcterms.referencesE. Mascioli, L. Leader, E. Flores, S. Trimbo, B. Bistrian, and G. Blackburn, “Enhanced survival to endotoxin in guinea pigs fed IV fish oil emulsion,” Lipids, vol. 23, no. 6, pp. 623–625, 1988.en
dcterms.referencesB. Di Luccia et al., “Rescue of fructose-induced metabolic syndrome by antibiotics or faecal transplantation in a rat model of obesity,” PLoS One, vol. 10, no. 8, p. e0134893, 2015en
dcterms.referencesI. Bergheim et al., “Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin,” J. Hepatol., vol. 48, no. 6, pp. 983–992, 2008en
dcterms.referencesX. Ouyang et al., “Fructose consumption as a risk factor for non-alcoholic fatty liver disease,” J. Hepatol., vol. 48, no. 6, pp. 993–999, 2008.en
dcterms.referencesA. Salonen and W. M. de Vos, “Impact of diet on human intestinal microbiota and health.,” Annu. Rev. Food Sci. Technol., vol. 5, pp. 239–62, 2014.en
dcterms.referencesH. Flint, K. Scott, P. Louis, and S. Duncan, “The role of the gut microbiota in nutrition and health,” Nat. Rev. Gastroenterol. Hepatol., vol. 9, no. 10, pp. 577–589, 2012.en
dcterms.referencesB. Ling, F. Peng, J. Alcorn, K. Lohmann, B. Bandy, and G. a Zello, “D-Lactate altered mitochondrial energy production in rat brain and heart but not liver,” Nutr. Metab. (Lond)., vol. 9, no. 1, p. 6, 2012.en
dcterms.referencesJ. B. Ewaschuk, J. M. Naylor, and G. a Zello, “D-lactate in human and ruminant metabolism.,” J. Nutr., vol. 135, no. 7, pp. 1619–1625, 2005.en
dcterms.referencesJ. R. Sheedy et al., “Increased D-lactic acid intestinal bacteria in patients with chronic fatigue syndrome,” In Vivo (Brooklyn)., vol. 23, no. 4, pp. 621–628, 2009.en
dcterms.referencesJ. Uribarri, M. S. Oh, and H. J. Carroll, “D-lactic acidosis. A review of clinical presentation, biochemical features, and pathophysiologic mechanisms.,” Medicine (Baltimore)., vol. 77, no. 2, pp. 73–82, 1998.en
dcterms.referencesK. J. Atkinson and R. K. Rao, “Role of protein tyrosine phosphorylation in acetaldehyde-induced disruption of epithelial tight junctions.,” Am. J. Physiol. Gastrointest. Liver Physiol., vol. 280, no. 6, pp. G1280–G1288, 2001.en
dcterms.referencesM. Salaspuro, “Bacteriocolonic pathway for ethanol oxidation: characteristics and implications,” Ann Med, vol. 28, no. 3, pp. 195–200, 1996.en
dcterms.referencesR. M. Wright, J. L. McManaman, and J. E. Repine, “Alcohol-induced breast cancer: A proposed mechanism,” Free Radic. Biol. Med., vol. 26, no. 3–4, pp. 348–354, 1999.en
dcterms.referencesG. den Besten, K. van Eunen, A. Groen, K. Venema, D. Reijngoud, and B. Bakker, “The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism,” J. Lipid Res., vol. 54, no. 9, pp. 2325–2340, 2013.en
dcterms.referencesR. Hughes, E. a Magee, and S. Bingham, “Protein degradation in the large intestine: relevance to colorectal cancer.,” Curr. Issues Intest. Microbiol., vol. 1, no. 2, pp. 51–58, 2000.en
dcterms.referencesW. R. Russell et al., “High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health,” Am. J. Clin. Nutr., vol. 93, no. 5, pp. 1062–1072, 2011.en
dcterms.referencesR. Koeth, Z. Wang, B. Levison, and J. Buffa, “Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis,” Nat. Med., vol. 19, no. 5, pp. 576–585, 2013.en
dcterms.referencesS. Bengmark, “Bio-ecological control of chronic liver disease and encephalopathy,” Metabolic Brain Disease, vol. 24, no. 1. pp. 223–236, 2009.en
dcterms.referencesR. Wade and C. Castro, “Redox reactivity of iron (III) porphyrins and heme proteins with nitric oxide. Nitrosyl transfer to carbon, oxygen, nitrogen and sulfur,” Chem. Res. Toxicol., vol. 3, no. 4, pp. 289–291, 1990en
dcterms.referencesG. R. Gibson, J. H. Cummings, and G. T. Macfarlane, “Growth and activities of sulphate-reducing bacteria in gut contents of healthy subjects and patients with ulcerative colitis,” FEMS Microbiol. Lett., vol. 86, no. 2, pp. 103–111, 1991.en
dcterms.referencesM. S. Attene-Ramos, E. D. Wagner, M. J. Plewa, and H. R. Gaskins, “Evidence that hydrogen sulfide is a genotoxic agent,” Mol Cancer Res, vol. 4, no. 1, pp. 9–14, 2006.en
dcterms.referencesR. J. Bloomer and K. H. Fisher-Wellman, “Lower postprandial oxidative stress in women compared with men,” Gend. Med., vol. 7, no. 4, pp. 340–349, 2010.en
dcterms.references] R. Caesar, F. Fåk, and F. Bäckhed, “Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism: Review,” J. Intern. Med., vol. 268, no. 4, pp. 320–328, 2010en
dcterms.referencesH. van der Vaart, D. S. Postma, W. Timens, and N. H. T. ten Hacken, “Acute effects of cigarette smoke on inflammation and oxidative stress: a review.,” Thorax, vol. 59, no. 8, pp. 713–721, 2004.en
dcterms.referencesX. Zhang, G. Zhang, H. Zhang, M. Karin, H. Bai, and D. Cai, “Hypothalamic IKKa/NF-kB and ER Stress Link Overnutrition to Energy Imbalance and Obesity,” Cell, vol. 135, no. 1, pp. 61–73, 2008.en
dcterms.referencesP. Sheth, S. Basuroy, C. Li, A. P. Naren, and R. K. Rao, “Role of phosphatidylinositol 3-kinase in oxidative stress-induced disruption of tight junctions.,” J. Biol. Chem., vol. 278, no. 49, pp. 49239–49245, 2003.en
dcterms.referencesM. Maes and J. Leunis, “Normalization of leaky gut in chronic fatigue syndrome (CFS) is accompanied by a clinical improvement: effects of age, duration of illness and the translocation,” Neuroendocrinol. Lett., vol. 29, no. 6, p. 902, 2008.en
dcterms.referencesM. L. Jones, C. J. Martoni, J. G. Ganopolsky, A. Labbé, and S. Prakash, “The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention.,” Expert Opin. Biol. Ther., vol. 14, no. 4, pp. 467–82, 2014.en
dcterms.referencesL. Furuya-Kanamori et al., “Comorbidities, Exposure to Medications, and the Risk of Community-Acquired Clostridium difficile Infection: A Systematic Review and Meta-analysis,” Infect. Control Hosp. Epidemiol., vol. 36, no. 02, pp. 132–141, 2014.en
dcterms.referencesR. Ungaro et al., “Antibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysis.,” Am. J. Gastroenterol., vol. 109, no. 11, pp. 1728–38, 2014.en
dcterms.referencesL. M. Cox and M. J. Blaser, “Antibiotics in early life and obesity.,” Nat. Rev. Endocrinol., vol. 11, no. 3, pp. 182–90, 2015.en
dcterms.referencesM. J. Blaser and S. Falkow, “What are the consequences of the disappearing human microbiota?,” Nat. Rev. Microbiol., vol. 7, no. 12, pp. 887–894, 2009.en
dcterms.referencesA. Moeller et al., “Cospeciation of gut microbiota with hominids,” Science (80-. )., vol. 353, no. 6297, pp. 380–382, 2016.en
dcterms.referencesK. E. Nichol, W. W. Poon, A. I. Parachikova, D. H. Cribbs, C. G. Glabe, and C. W. Cotman, “Exercise alters the immune profile in Tg2576 Alzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid.,” J. Neuroinflammation, vol. 5, no. 1, p. 13, 2008en
dcterms.referencesR. Starkie, S. R. Ostrowski, S. Jauffred, M. Febbraio, and B. K. Pedersen, “Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans.,” FASEB J., vol. 17, no. 8, pp. 884–886, 2003.en
dcterms.referencesK. H. Schmitz, D. R. Jacobs, C.-P. Hong, J. Steinberger, a Moran, and a R. Sinaiko, “Association of physical activity with insulin sensitivity in children.,” Int. J. Obes. Relat. Metab. Disord., vol. 26, no. 10, pp. 1310–1316, 2002.en
dcterms.referencesM. Matsumoto et al., “Impact of intestinal microbiota on intestinal luminal metabolome,” Sci. Rep., vol. 2, p. 233, 2012.en
dcterms.referencesR. Campos-Rodriguez, M. Godinez-Victoria, A. A. Arciniega-Martínez, I.M., Resendiz-Albor, H. Reyna-Garfias, T. R. Cruz-Hernández, and M. E. Drago-Serrano, “Protective Effect of Moderate Exercise for BALB/c Mice with Salmonella Typhimurium Infection.,” Int. J. Sports Med., vol. 37, no. 01, pp. 63–70, 2016.en
dcterms.referencesH. Sies, W. Stahl, and A. Sevanian, “Nutritional, dietary and postprandial oxidative stress,” J. Nutr., vol. 135, no. 5, pp. 969–972, 2005.en
dcterms.referencesK. Wang, X. Jin, Y. Chen, Z. Song, and X. Jiang, “Polyphenol-Rich Propolis Extracts Strengthen Intestinal Barrier Function by Activating AMPK and ERK Signaling,” Nutrients, vol. 8, no. 5, p. 272, 2016en
dcterms.referencesT. Cowan, M. Palmnas, K. Ardell, and J. Yang, “Chronic coffee consumption alters gut microbiome: potential mechanism to explain the protective effects of coffee on type 2 diabetes?,” FASEB J., vol. 27, no. 1_MeetingAbstracts, pp. 951–1, 2013.en
dcterms.referencesF. F. Anhê et al., “A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice.,” Gut, vol. 64, no. 6, pp. 872–83, 2015.en
dcterms.referencesH.-J. He, G.-Y. Wang, Y. Gao, W.-H. Ling, Z.-W. Yu, and T.-R. Jin, “Curcumin attenuates Nrf2 signaling defect, oxidative stress in muscle and glucose intolerance in high fat diet-fed mice,” World J. Diabetes, vol. 3, no. 5, p. 94, 2012.en
dcterms.referencesS. Ghosh et al., “Fish Oil Attenuates Omega-6 Polyunsaturated Fatty Acid-Induced Dysbiosis and Infectious Colitis but Impairs LPS Dephosphorylation Activity Causing Sepsis,” PLoS One, vol. 8, no. 2, p. e55468, 2013en
dcterms.referencesD. Roopchand, R. Carmody, and P. Kuhn, “Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphila and Attenuate High-Fat Diet–Induced Metabolic Syndrome,” Diabetes, vol. 64, no. 8, pp. 2847–2858, 2015.en
dcterms.referencesL. Thompson and R. C. Spiller, “Impact of polyunsaturated fatty acids on human colonic bacterial metabolism: an in vitro and in vivo study.,” Br. J. Nutr., vol. 74, no. 5, pp. 733–41, 1995.en
dcterms.referencesFAO/WHO, “Guidelines for the evaluation of probiotics in food,” London World Heal. Organ. ON, Canada Food Agric. Organ., 2002.en
dcterms.referencesS. Doron and S. L. Gorbach, “Probiotics: their role in the treatment and prevention of disease.,” Expert Rev. Anti. Infect. Ther., vol. 4, no. 2, pp. 261–275, 2006.en
dcterms.referencesD. Y. Park et al., “Supplementation of Lactobacillus curvatus KY1032 in Diet-Induced Obese Mice Is Associated with Gut Microbial Changes and Reduction in Obesity,” PLoS One, vol. 8, no. 3, p. 59470, 2013.en
dcterms.referencesD. Y. Park, Y. T. Ahn, C. S. Huh, R. A. Mcgregor, and M. S. Choi, “Dual probiotic strains suppress high fructoseinduced metabolic syndrome,” World J. Gastroenterol., vol. 19, no. 2, pp. 274–283, 2013.en
dcterms.referencesA. Everard et al., “Cross-talk between Akkermansia muciniphila and intestinal epithelium controls dietinduced obesity.,” Proc. Natl. Acad. Sci. U. S. A., vol. 110, no. 22, pp. 9066–71, 2013.en
dcterms.referencesP. G. Cano, A. Santacruz, A. Moya, and Y. Sanz, “Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity,” PLoS One, vol. 7, no. 7, p. e41079, 2012.en
dcterms.referencesB. R. Goldin and S. L. Gorbach, “Alterations of the intestinal microflora by diet, oral antibiotics, and lactobacillus: Decreased production of free amines from aromatic nitro compounds, azo dyes, and glucuronides,” J. Natl. Cancer Inst., vol. 73, no. 3, pp. 689–695, 1984en
dcterms.referencesB. R. Goldin and S. L. Gorbach, “The effect of milk and lactobacillus feeding on human intestinal bacterial enzyme activity,” Am. J. Clin. Nutr., vol. 39, no. 5, pp. 756–761, 1984.en
dcterms.referencesI. A. Kirpich et al., “Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: a pilot study,” Alcohol, vol. 42, no. 8, pp. 675–682, 2008.en
dcterms.referencesY. Wang et al., “Lactobacillus rhamnosus GG treatment potentiates intestinal hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury,” Am. J. Pathol., vol. 179, no. 6, pp. 2866–2875, 2011en
dcterms.referencesM. Naruszewicz, M.-L. Johansson, D. Zapolska-Downar, and H. Bukowska, “Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers.,” Am. J. Clin. Nutr., vol. 76, no. 6, pp. 1249–55, 2002.en
dcterms.referencesI. Kimura et al., “The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43.,” Nat. Commun., vol. 4, p. 1829, 2013.en
dcterms.referencesK.-Y. Park, B. Kim, and C.-K. Hyun, “Lactobacillus rhamnosus GG improves glucose tolerance through alleviating ER stress and suppressing macrophage activation in db/db mice.,” J. Clin. Biochem. Nutr., vol. 56, no. 3, pp. 240–6, 2015.en
dcterms.referencesK. A. Tappenden, D. M. Albin, A. L. Bartholome, and H. F. Mangian, “Glucagon-like peptide-2 and short-chain fatty acids: a new twist to an old story,” J Nutr, vol. 133, no. 11, pp. 3717–3720, 2003.en
dcterms.referencesV. Marcil, E. Delvin, C. Garofalo, and E. Levy, “Butyrate impairs lipid transport by inhibiting microsomal triglyceride transfer protein in Caco-2 cells.,” J. Nutr., vol. 133, no. 7, pp. 2180–2183, 2003.en
dcterms.referencesN. M. Delzenne, A. M. Neyrinck, and P. D. Cani, “Gut microbiota and metabolic disorders: How prebiotic can work?,” Br. J. Nutr., vol. 109 Suppl, pp. S81-5, 2013.en
dcterms.referencesM. Galisteo, J. Duarte, and A. Zarzuelo, “Effects of dietary fibers on disturbances clustered in the metabolic syndrome,” Journal of Nutritional Biochemistry, vol. 19, no. 2. pp. 71–84, 2008.en
dcterms.referencesE. Chambers et al., “Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults,” Gut, p. gutjnl, 2014.en
dcterms.referencesG. Frost, D. Morrison, and T. Preston, “Compounds And Their Effects On Appetite Control And Insulin Sensitivity,” 2013.en
dcterms.referencesY.-Y. Ma, L. Li, C. Yu, Z. Shen, L. Chen, and Y. Li, “Effects of probiotics on nonalcoholic fatty liver disease: a meta-analysis.,” World J. Gastroenterol., vol. 19, no. 40, pp. 6911–8, 2013en
dcterms.referencesQ. Liu, Z. P. Duan, D. K. Ha, S. Bengmark, J. Kurtovic, and S. M. Riordan, “Synbiotic Modulation of Gut Flora: Effect on Minimal Hepatic Encephalopathy in Patients with Cirrhosis,” Hepatology, vol. 39, no. 5, pp. 1441– 1449, 2004.en
dcterms.referencesZ. Asemi, A. Khorrami-Rad, and S. Alizadeh, “Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial,” Clin. Nutr., vol. 33, no. 2, pp. 198–203, 2014. -en
dcterms.referencesY. Kadooka et al., “Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial,” Eur. J. Clin. Nutr., vol. 64, no. 10, pp. 636–643, 2010.en
dcterms.referencesL. V. McFarland, “Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease,” Am. J. Gastroenterol., vol. 101, no. 4, pp. 812–822, 2006.en
dcterms.references] M. Ito, K. Ohishi, Y. Yoshida, W. Yokoi, and H. Sawada, “Antioxidative effects of lactic acid bacteria on the colonic mucosa of iron-overloaded mice,” J. Agric. Food Chem., vol. 51, no. 15, pp. 4456–4460, 2003.en
dcterms.referencesT. Kullisaar, J. Shepetova, and K. Zilmer, “An antioxidant probiotic reduces postprandial lipemia and oxidative stress,” Cent. Eur. J. Biol., vol. 6, no. 1, pp. 32–40, 2011.en
dcterms.referencesH. Chung, J. Yu, I. Lee, and M. Liu, “Intestinal removal of free fatty acids from hosts by Lactobacilli for the treatment of obesity,” FEBS Open Bio, vol. 6, no. 1, pp. 64–76, 2016.en
dcterms.referencesS. Thomas, D. Metzke, J. Schmitz, Y. Dörffel, and D. C. Baumgart, “Anti-inflammatory effects of Saccharomyces boulardii mediated by myeloid dendritic cells from patients with Crohn’s disease and ulcerative colitis.,” Am. J. Physiol. Gastrointest. Liver Physiol., vol. 301, no. 6, pp. G1083-92, 2011.en
dcterms.referencesJ. P. Buts, N. Dekeyser, C. Stilmant, E. Delem, F. Smets, and E. Sokal, “Saccharomyces boulardii produces in rat small intestine a novel protein phosphatase that inhibits Escherichia coli endotoxin by dephosphorylation,” Pediatr. Res., vol. 60, no. 1, pp. 24–29, 2006.en
dcterms.referencesM. Fujiya et al., “The Bacillus subtilis Quorum-Sensing Molecule CSF Contributes to Intestinal Homeostasis via OCTN2, a Host Cell Membrane Transporter,” Cell Host Microbe, vol. 1, no. 4, pp. 299–308, 2007.en
dcterms.referencesA. Seth, F. Yan, D. Polk, and R. Rao, “Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruption by a PKC-and MAP kinase-dependent mechanism,” Am. J. Physiol. Liver Physiol., vol. 294, no. 4, pp. G1060–G1069, 2008.en
dcterms.referencesE. O. Petrof et al., “Probiotics inhibit nuclear factor-kB and induce heat shock proteins in colonic epithelial cells through proteasome inhibition,” Gastroenterology, vol. 127, no. 5, pp. 1474–1487, 2004.en
dcterms.referencesA. Drewnowski and B. M. Popkin, “The Nutrition Transition: New Trends in the Global Diet,” Nutr. Rev., vol. 55, no. 2, pp. 31–43, 1997.eng
dcterms.referencesF. B. Hu et al., “Reproducibility and validity of dietary patterns assessed with a food-frequency questionnaire,” Am. J. Clin. Nutr., vol. 69, no. 2, pp. 243–249, 1999.eng
dcterms.referencesM. L. Slattery, K. M. Boucher, B. J. Caan, J. D. Potter, and K. N. Ma, “Eating patterns and risk of colon cancer.,” Am. J. Epidemiol., vol. 148, no. 1, pp. 4–16, 1998.eng
dcterms.referencesA. Nanri et al., “Reproducibility and validity of dietary patterns assessed by a food frequency questionnaire used in the 5-year follow-up survey of the Japan Public Health Center-Based Prospective Study,” J. Epidemiol., vol. 22, no. 3, pp. 205–215, 2012eng
dcterms.referencesL. Cordain et al., “Origins and evolution of the Western diet: health implications for the 21st century.,” Am. J. Clin. Nutr., vol. 81, no. 2, pp. 341–54, 2005.eng
dcterms.referencesW. Price, “Nutrition and Physical Degeneration,” Can. Med. Assoc. J., vol. 42, no. 2, p. 208, 1939.eng
dcterms.referencesS. Parry and L. Straker, “The contribution of office work to sedentary behaviour associated risk,” BMC Public Health, vol. 13, no. 1, pp. 1–10, 2013.eng
dcterms.referencesCDC, “Vital signs: binge drinking prevalence, frequency, and intensity among adults - United States, 2010.,” 2012.eng
dcterms.referencesWHO, “Global status report on noncommunicable diseases 2014,” World Health, p. 176, 2014.eng
dcterms.referencesS. Kanoski and T. Davidson, “Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity,” Physiol. Behav., vol. 103, no. 1, pp. 59–68, 2011.eng
dcterms.referencesL. Bazzano, “Dietary intake of fruit and vegetables and risk of diabetes mellitus and cardiovascular diseases,” Geneva WHO, 2005.eng
dcterms.referencesF. M. Trovato, D. Catalano, G. F. Martines, P. Pace, and G. M. Trovato, “Mediterranean diet and non-alcoholic fatty liver disease. The need of extended and comprehensive interventions,” Clin. Nutr., vol. 34, no. 1, pp. 86–88, 2015.eng
dcterms.referencesJ. M. Geleijnse, F. J. Kok, and D. E. Grobbee, “Impact of dietary and lifestyle factors on the prevalence of hypertension in Western populations,” Eur. J. Public Health, vol. 14, no. 3, pp. 235–239, 2004.eng
dcterms.referencesS. Jehle, A. Zanetti, J. Muser, H. N. Hulter, and R. Krapf, “Partial neutralization of the acidogenic Western diet with potassium citrate increases bone mass in postmenopausal women with osteopenia.,” J. Am. Soc. Nephrol., vol. 17, no. 11, pp. 3213–3222, 2006.eng
dcterms.referencesA. Manzel, D. N. Muller, D. A. Hafler, S. E. Erdman, R. A. Linker, and M. Kleinewietfeld, “Role of ‘western diet’ in inflammatory autoimmune diseases,” Curr. Allergy Asthma Rep., vol. 14, no. 1, pp. 1–8, 2014.eng
dcterms.referencesH. Adlercreutz, “Western diet and Western diseases: Some hormonal and biochemical mechanisms and associations,” Scand. J. Clin. Lab. Invest. Suppl., vol. 50, no. sup201, pp. 3–23, 1990.eng
dcterms.referencesJ. Lederberg and A. McCray, “Ome SweetOmics--A Genealogical Treasury of Words,” Sci., vol. 15, no. 7, p. 8, 2001.eng
dcterms.referencesC. Huttenhower et al., “Structure, function and diversity of the healthy human microbiome,” Nature, vol. 486, no. 7402, pp. 207–214, 2012.eng
dcterms.referencesP. B. Eckburg et al., “Diversity of the human intestinal microbial flora.,” Science, vol. 308, no. 5728, pp. 1635– 8, 2005eng
dcterms.referencesJ. Qin, R. Li, J. Raes, and M. Arumugam, “A human gut microbial gene catalogue established by metagenomic sequencing,” Nature, vol. 464, no. 7285, pp. 59–65, 2010.eng
dcterms.referencesM. Frémont, D. Coomans, S. Massart, and K. De Meirleir, “High-throughput 16S rRNA gene sequencing reveals alterations of intestinal microbiota in myalgic encephalomyelitis/chronic fatigue syndrome patients,” Anaerobe, vol. 22, pp. 50–56, 2013.eng
dcterms.referencesX. Zhang et al., “The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment.,” Nat. Med., vol. 21, no. 8, pp. 895–905, 2015.eng
dcterms.referencesC. T. Brown et al., “Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes,” PLoS One, vol. 6, no. 10, 2011.eng
dcterms.referencesN. Larsen et al., “Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults,” PLoS One, vol. 5, no. 2, 2010.eng
dcterms.referencesK. Forslund et al., “Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota.,” Nature, vol. 528, no. 7581, pp. 262–266, 2015.eng
dcterms.referencesM. Joossens et al., “Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives,” Gut, vol. 60, no. 5, pp. 631–637, 2011.eng
dcterms.referencesA. R. Erickson et al., “Integrated Metagenomics/Metaproteomics Reveals Human Host-Microbiota Signatures of Crohn’s Disease,” PLoS One, vol. 7, no. 11, 2012.eng
dcterms.referencesI. A. Finnie, A. D. Dwarakanath, B. A. Taylor, and J. M. Rhodes, “Colonic mucin synthesis is increased by sodium butyrate.,” Gut, vol. 36, no. 1, pp. 93–9, 1995.eng
dcterms.referencesL. Peng, Z.-R. Li, R. S. Green, I. R. Holzman, and J. Lin, “Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers.,” J. Nutr., vol. 139, no. 9, pp. 1619–1625, 2009.eng
dcterms.referencesM. A. R. Vinolo, H. G. Rodrigues, R. T. Nachbar, and R. Curi, “Regulation of Inflammation by Short Chain Fatty Acids,” Nutrients, vol. 3, no. 12, pp. 858–876, 2011.eng
dcterms.referencesP. Louis, P. Young, G. Holtrop, and H. J. Flint, “Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene,” Environ. Microbiol., vol. 12, no. 2, pp. 304–314, 2010.eng
dcterms.referencesM. J. Hopkins, R. Sharp, and G. T. Macfarlane, “Age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles.,” Gut, vol. 48, no. 2, pp. 198–205, 2001.eng
dcterms.referencesP. J. Turnbaugh et al., “A core gut microbiome in obese and lean twins.,” Nature, vol. 457, no. 7228, pp. 480– 484, 2009.eng
dcterms.referencesL. C. Kong et al., “Gut microbiota after gastric bypass in human obesity: Increased richness and associations of bacterial genera with adipose tissue genes,” Am. J. Clin. Nutr., vol. 98, no. 1, pp. 16–24, 2013.eng
dcterms.referencesC. de Weerth, S. Fuentes, P. Puylaert, and W. M. de Vos, “Intestinal microbiota of infants with colic: development and specific signatures.,” Pediatrics, vol. 131, no. 2, pp. e550-8, 2013.eng
dcterms.references] T. Yatsunenko et al., “Human gut microbiome viewed across age and geography,” Nature, vol. 486, no. 7402, pp. 222–227, 2012.eng
dcterms.referencesC. A. Lozupone, M. E. Rhodes, C. P. Neff, A. P. Fontenot, T. B. Campbell, and B. E. Palmer, “HIV-induced alteration in gut microbiota,” Gut Microbes, vol. 5, no. 4, pp. 562–570, 2014eng
dcterms.referencesW. H. W. Tang et al., “Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk.,” N. Engl. J. Med., vol. 368, no. 17, pp. 1575–84, 2013.eng
dcterms.referencesR. a Koeth et al., “Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis,” Nat. Med., vol. 19, no. April, pp. 576–85, 2013.eng
dcterms.referencesT. Poutahidis et al., “Microbial Reprogramming Inhibits Western Diet-Associated Obesity,” PLoS One, vol. 8, no. 7, p. e68596, 2013.eng
dcterms.referencesF. Armougom, M. Henry, B. Vialettes, D. Raccah, and D. Raoult, “Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients,” PLoS One, vol. 4, no. 9, p. e7125, 2009.eng
dcterms.referencesM. Million et al., “Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii.,” Int. J. Obes. (Lond)., vol. 36, no. 6, pp. 817–25, 2012.eng
dcterms.referencesF. Fåk and F. Bäckhed, “Lactobacillus reuteri prevents diet-induced obesity, but not atherosclerosis, in a strain dependent fashion in Apoe−/− mice,” PLoS One, vol. 7, no. 10, p. e46837, 2012.eng
dcterms.referencesM. P. Diaz-Ropero et al., “Two Lactobacillus strains, isolated from breast milk, differently modulate the immune response,” J. Appl. Microbiol., vol. 102, no. 2, pp. 337–343, 2007.eng
dcterms.referencesB. T. Burton and W. R. Foster, “Health implications of obesity: an NIH Consensus Development Conference,” J. Am. Diet. Assoc., vol. 85, no. 9, pp. 1117–1121, 1985eng
dcterms.referencesC. Ogden, S. Yanovski, M. Carroll, and K. Flegal, “The epidemiology of obesity,” Gastroenterology, vol. 132, no. 6, pp. 2087–2102, 2007.eng
dcterms.referencesJ. V Van Vliet-Ostaptchouk et al., “The prevalence of Metabolic Syndrome and metabolically healthy obesity in Europe: a collaborative analysis of ten large cohort studies.,” BMC Endocr. Disord., vol. 14, no. 9, pp. 1– 13, 2014.eng
dcterms.referencesN. Klöting et al., “Insulin-sensitive obesity,” Am. J. Physiol. - Endocrinol. Metab., vol. 299, no. 3, pp. 506–515, 2010.eng
dcterms.referencesJ. Naukkarinen et al., “Characterising metabolically healthy obesity in weight-discordant monozygotic twins,” Diabetologia, vol. 57, no. 1, pp. 167–176, 2014.eng
dcterms.referencesK. Azuma et al., “Higher liver fat content among Japanese in Japan compared with non-Hispanic whites in the United States,” Metabolism., vol. 58, no. 8, pp. 1200–1207, 2009.eng
dcterms.referencesR. W. Walker et al., “High rates of fructose malabsorption are associated with reduced liver fat in obese African Americans.,” J. Am. Coll. Nutr., vol. 31, no. 5, pp. 369–74, 2012eng
dcterms.referencesR. Guerrero, G. L. Vega, S. M. Grundy, and J. D. Browning, “Ethnic differences in hepatic steatosis: An insulin resistance paradox?,” Hepatology, vol. 49, no. 3, pp. 791–801, 2009.eng
dcterms.referencesM. G. Marmot, S. L. Syme, A. Kagan, H. Kato, J. B. Cohen, and J. Belsky, “Epidemiologic studies of coronary heart disease and stroke in Japanese men living in Japan, Hawaii and California: prevalence of coronary and hypertensive heart disease and associated risk factors.,” Am. J. Epidemiol., vol. 102, no. 6, pp. 514–25, 1975.eng
dcterms.referencesC. B. Newgard, “Interplay between lipids and branched-chain amino acids in development of insulin resistance,” Cell Metabolism, vol. 15, no. 5. pp. 606–614, 2012.eng
dcterms.referencesM. Raman et al., “Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease,” Clin. Gastroenterol. Hepatol., vol. 11, no. 7, pp. 868–875, 2013.eng
dcterms.referencesL. Zhu et al., “Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH,” Hepatology, vol. 57, no. 2, pp. 601–609, 2013.eng
dcterms.referencesJ. Boursier and A. M. Diehl, “Nonalcoholic Fatty Liver Disease and the Gut Microbiome,” Clinics in Liver Disease, vol. 20, no. 2. pp. 263–275, 2016.eng
dcterms.referencesF. Bäckhed, J. K. Manchester, C. F. Semenkovich, and J. I. Gordon, “Mechanisms underlying the resistance to diet-induced obesity in germ-free mice,” Proc. Natl. Acad. Sci., vol. 104, no. 3, pp. 979–984, 2007.eng
dcterms.referencesA. M. W. Petersen and B. K. Pedersen, “The anti-inflammatory effect of exercise.,” J. Appl. Physiol., vol. 98, no. 4, pp. 1154–1162, 2005.eng
dcterms.referencesJ. M. Wells, O. Rossi, M. Meijerink, and P. van Baarlen, “Epithelial crosstalk at the microbiota-mucosal interface.,” Proc. Natl. Acad. Sci. U. S. A., vol. 108 Suppl, pp. 4607–4614, 2011.eng
dcterms.referencesG. G. Muccioli et al., “The endocannabinoid system links gut microbiota to adipogenesis.,” Mol. Syst. Biol., vol. 6, no. 392, p. 392, 2010.eng
dcterms.referencesH. Nakarai et al., “Adipocyte-macrophage interaction may mediate LPS-induced low-grade inflammation: potential link with metabolic complications.,” Innate Immun., vol. 18, no. 1, pp. 164–70, 2012.eng
dcterms.referencesF. S. Lira et al., “Endotoxin levels correlate positively with a sedentary lifestyle and negatively with highly trained subjects.,” Lipids Health Dis., vol. 9, no. 1, p. 82, 2010.eng
dcterms.referencesE. Pace et al., “Cigarette smoke increases Toll-like receptor 4 and modifies lipopolysaccharide-mediated responses in airway epithelial cells,” Immunology, vol. 124, no. 3, pp. 401–411, 2008eng
dcterms.referencesM. Lyte and S. Ernst, “Catecholamine induced growth of gram negative bacteria,” Life Sci., vol. 50, no. 3, pp. 203–212, 1992.eng
dcterms.referencesP. P. E. Freestone, S. M. Sandrini, R. D. Haigh, and M. Lyte, “Microbial endocrinology: how stress influences susceptibility to infection,” Trends in Microbiology, vol. 16, no. 2. pp. 55–64, 2008eng
dcterms.referencesM. Lyte and M. T. Bailey, “Neuroendocrine-bacterial interactions in a neurotoxin-induced model of trauma.,” J. Surg. Res., vol. 70, no. 2, pp. 195–201, 1997.eng
dcterms.referencesM. Lyte, C. D. Frank, and B. T. Green, “Production of an autoinducer of growth by norepinephrine cultured Escherichia coli O157:H7,” FEMS Microbiol. Lett., vol. 139, no. 2–3, pp. 155–159, 1996.eng
dcterms.referencesP. D. Cani et al., “Metabolic endotoxemia initiates obesity and insulin resistance,” Diabetes, vol. 56, no. 7, pp. 1761–1772, 2007.eng
dcterms.referencesS. Ghoshal, J. Witta, J. Zhong, W. de Villiers, and E. Eckhardt, “Chylomicrons promote intestinal absorption of lipopolysaccharides.,” J. Lipid Res., vol. 50, no. 1, pp. 90–97, 2009.eng
dcterms.referencesS. Sadeghi, F. A. Wallace, and P. C. Calder, “Dietary lipids modify the cytokine response to bacterial lipopolysaccharide in mice,” Immunology, vol. 96, no. 3, pp. 404–410, 1999.eng
dcterms.referencesE. Mascioli, L. Leader, E. Flores, S. Trimbo, B. Bistrian, and G. Blackburn, “Enhanced survival to endotoxin in guinea pigs fed IV fish oil emulsion,” Lipids, vol. 23, no. 6, pp. 623–625, 1988.eng
dcterms.referencesB. Di Luccia et al., “Rescue of fructose-induced metabolic syndrome by antibiotics or faecal transplantation in a rat model of obesity,” PLoS One, vol. 10, no. 8, p. e0134893, 2015eng
dcterms.referencesI. Bergheim et al., “Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin,” J. Hepatol., vol. 48, no. 6, pp. 983–992, 2008eng
dcterms.referencesX. Ouyang et al., “Fructose consumption as a risk factor for non-alcoholic fatty liver disease,” J. Hepatol., vol. 48, no. 6, pp. 993–999, 2008.eng
dcterms.referencesA. Salonen and W. M. de Vos, “Impact of diet on human intestinal microbiota and health.,” Annu. Rev. Food Sci. Technol., vol. 5, pp. 239–62, 2014.eng
dcterms.referencesH. Flint, K. Scott, P. Louis, and S. Duncan, “The role of the gut microbiota in nutrition and health,” Nat. Rev. Gastroenterol. Hepatol., vol. 9, no. 10, pp. 577–589, 2012.eng
dcterms.referencesB. Ling, F. Peng, J. Alcorn, K. Lohmann, B. Bandy, and G. a Zello, “D-Lactate altered mitochondrial energy production in rat brain and heart but not liver,” Nutr. Metab. (Lond)., vol. 9, no. 1, p. 6, 2012.eng
dcterms.referencesJ. B. Ewaschuk, J. M. Naylor, and G. a Zello, “D-lactate in human and ruminant metabolism.,” J. Nutr., vol. 135, no. 7, pp. 1619–1625, 2005.eng
dcterms.referencesJ. R. Sheedy et al., “Increased D-lactic acid intestinal bacteria in patients with chronic fatigue syndrome,” In Vivo (Brooklyn)., vol. 23, no. 4, pp. 621–628, 2009.eng
dcterms.referencesJ. Uribarri, M. S. Oh, and H. J. Carroll, “D-lactic acidosis. A review of clinical presentation, biochemical features, and pathophysiologic mechanisms.,” Medicine (Baltimore)., vol. 77, no. 2, pp. 73–82, 1998.eng
dcterms.referencesK. J. Atkinson and R. K. Rao, “Role of protein tyrosine phosphorylation in acetaldehyde-induced disruption of epithelial tight junctions.,” Am. J. Physiol. Gastrointest. Liver Physiol., vol. 280, no. 6, pp. G1280–G1288, 2001.eng
dcterms.referencesM. Salaspuro, “Bacteriocolonic pathway for ethanol oxidation: characteristics and implications,” Ann Med, vol. 28, no. 3, pp. 195–200, 1996.eng
dcterms.referencesR. M. Wright, J. L. McManaman, and J. E. Repine, “Alcohol-induced breast cancer: A proposed mechanism,” Free Radic. Biol. Med., vol. 26, no. 3–4, pp. 348–354, 1999.eng
dcterms.referencesG. den Besten, K. van Eunen, A. Groen, K. Venema, D. Reijngoud, and B. Bakker, “The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism,” J. Lipid Res., vol. 54, no. 9, pp. 2325–2340, 2013.eng
dcterms.referencesR. Hughes, E. a Magee, and S. Bingham, “Protein degradation in the large intestine: relevance to colorectal cancer.,” Curr. Issues Intest. Microbiol., vol. 1, no. 2, pp. 51–58, 2000.eng
dcterms.referencesW. R. Russell et al., “High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health,” Am. J. Clin. Nutr., vol. 93, no. 5, pp. 1062–1072, 2011.eng
dcterms.referencesR. Koeth, Z. Wang, B. Levison, and J. Buffa, “Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis,” Nat. Med., vol. 19, no. 5, pp. 576–585, 2013.eng
dcterms.referencesS. Bengmark, “Bio-ecological control of chronic liver disease and encephalopathy,” Metabolic Brain Disease, vol. 24, no. 1. pp. 223–236, 2009.eng
dcterms.referencesR. Wade and C. Castro, “Redox reactivity of iron (III) porphyrins and heme proteins with nitric oxide. Nitrosyl transfer to carbon, oxygen, nitrogen and sulfur,” Chem. Res. Toxicol., vol. 3, no. 4, pp. 289–291, 1990eng
dcterms.referencesG. R. Gibson, J. H. Cummings, and G. T. Macfarlane, “Growth and activities of sulphate-reducing bacteria in gut contents of healthy subjects and patients with ulcerative colitis,” FEMS Microbiol. Lett., vol. 86, no. 2, pp. 103–111, 1991.eng
dcterms.referencesM. S. Attene-Ramos, E. D. Wagner, M. J. Plewa, and H. R. Gaskins, “Evidence that hydrogen sulfide is a genotoxic agent,” Mol Cancer Res, vol. 4, no. 1, pp. 9–14, 2006.eng
dcterms.referencesR. J. Bloomer and K. H. Fisher-Wellman, “Lower postprandial oxidative stress in women compared with men,” Gend. Med., vol. 7, no. 4, pp. 340–349, 2010.eng
dcterms.references] R. Caesar, F. Fåk, and F. Bäckhed, “Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism: Review,” J. Intern. Med., vol. 268, no. 4, pp. 320–328, 2010eng
dcterms.referencesH. van der Vaart, D. S. Postma, W. Timens, and N. H. T. ten Hacken, “Acute effects of cigarette smoke on inflammation and oxidative stress: a review.,” Thorax, vol. 59, no. 8, pp. 713–721, 2004.eng
dcterms.referencesX. Zhang, G. Zhang, H. Zhang, M. Karin, H. Bai, and D. Cai, “Hypothalamic IKKa/NF-kB and ER Stress Link Overnutrition to Energy Imbalance and Obesity,” Cell, vol. 135, no. 1, pp. 61–73, 2008.eng
dcterms.referencesP. Sheth, S. Basuroy, C. Li, A. P. Naren, and R. K. Rao, “Role of phosphatidylinositol 3-kinase in oxidative stress-induced disruption of tight junctions.,” J. Biol. Chem., vol. 278, no. 49, pp. 49239–49245, 2003.eng
dcterms.referencesM. Maes and J. Leunis, “Normalization of leaky gut in chronic fatigue syndrome (CFS) is accompanied by a clinical improvement: effects of age, duration of illness and the translocation,” Neuroendocrinol. Lett., vol. 29, no. 6, p. 902, 2008.eng
dcterms.referencesM. L. Jones, C. J. Martoni, J. G. Ganopolsky, A. Labbé, and S. Prakash, “The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention.,” Expert Opin. Biol. Ther., vol. 14, no. 4, pp. 467–82, 2014.eng
dcterms.referencesL. Furuya-Kanamori et al., “Comorbidities, Exposure to Medications, and the Risk of Community-Acquired Clostridium difficile Infection: A Systematic Review and Meta-analysis,” Infect. Control Hosp. Epidemiol., vol. 36, no. 02, pp. 132–141, 2014.eng
dcterms.referencesR. Ungaro et al., “Antibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysis.,” Am. J. Gastroenterol., vol. 109, no. 11, pp. 1728–38, 2014.eng
dcterms.referencesL. M. Cox and M. J. Blaser, “Antibiotics in early life and obesity.,” Nat. Rev. Endocrinol., vol. 11, no. 3, pp. 182–90, 2015.eng
dcterms.referencesM. J. Blaser and S. Falkow, “What are the consequences of the disappearing human microbiota?,” Nat. Rev. Microbiol., vol. 7, no. 12, pp. 887–894, 2009.eng
dcterms.referencesA. Moeller et al., “Cospeciation of gut microbiota with hominids,” Science (80-. )., vol. 353, no. 6297, pp. 380–382, 2016.eng
dcterms.referencesK. E. Nichol, W. W. Poon, A. I. Parachikova, D. H. Cribbs, C. G. Glabe, and C. W. Cotman, “Exercise alters the immune profile in Tg2576 Alzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid.,” J. Neuroinflammation, vol. 5, no. 1, p. 13, 2008eng
dcterms.referencesR. Starkie, S. R. Ostrowski, S. Jauffred, M. Febbraio, and B. K. Pedersen, “Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans.,” FASEB J., vol. 17, no. 8, pp. 884–886, 2003.eng
dcterms.referencesK. H. Schmitz, D. R. Jacobs, C.-P. Hong, J. Steinberger, a Moran, and a R. Sinaiko, “Association of physical activity with insulin sensitivity in children.,” Int. J. Obes. Relat. Metab. Disord., vol. 26, no. 10, pp. 1310–1316, 2002.eng
dcterms.referencesM. Matsumoto et al., “Impact of intestinal microbiota on intestinal luminal metabolome,” Sci. Rep., vol. 2, p. 233, 2012.eng
dcterms.referencesR. Campos-Rodriguez, M. Godinez-Victoria, A. A. Arciniega-Martínez, I.M., Resendiz-Albor, H. Reyna-Garfias, T. R. Cruz-Hernández, and M. E. Drago-Serrano, “Protective Effect of Moderate Exercise for BALB/c Mice with Salmonella Typhimurium Infection.,” Int. J. Sports Med., vol. 37, no. 01, pp. 63–70, 2016.eng
dcterms.referencesH. Sies, W. Stahl, and A. Sevanian, “Nutritional, dietary and postprandial oxidative stress,” J. Nutr., vol. 135, no. 5, pp. 969–972, 2005.eng
dcterms.referencesK. Wang, X. Jin, Y. Chen, Z. Song, and X. Jiang, “Polyphenol-Rich Propolis Extracts Strengthen Intestinal Barrier Function by Activating AMPK and ERK Signaling,” Nutrients, vol. 8, no. 5, p. 272, 2016eng
dcterms.referencesT. Cowan, M. Palmnas, K. Ardell, and J. Yang, “Chronic coffee consumption alters gut microbiome: potential mechanism to explain the protective effects of coffee on type 2 diabetes?,” FASEB J., vol. 27, no. 1_MeetingAbstracts, pp. 951–1, 2013.eng
dcterms.referencesF. F. Anhê et al., “A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice.,” Gut, vol. 64, no. 6, pp. 872–83, 2015.eng
dcterms.referencesH.-J. He, G.-Y. Wang, Y. Gao, W.-H. Ling, Z.-W. Yu, and T.-R. Jin, “Curcumin attenuates Nrf2 signaling defect, oxidative stress in muscle and glucose intolerance in high fat diet-fed mice,” World J. Diabetes, vol. 3, no. 5, p. 94, 2012.eng
dcterms.referencesS. Ghosh et al., “Fish Oil Attenuates Omega-6 Polyunsaturated Fatty Acid-Induced Dysbiosis and Infectious Colitis but Impairs LPS Dephosphorylation Activity Causing Sepsis,” PLoS One, vol. 8, no. 2, p. e55468, 2013eng
dcterms.referencesD. Roopchand, R. Carmody, and P. Kuhn, “Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphila and Attenuate High-Fat Diet–Induced Metabolic Syndrome,” Diabetes, vol. 64, no. 8, pp. 2847–2858, 2015.eng
dcterms.referencesL. Thompson and R. C. Spiller, “Impact of polyunsaturated fatty acids on human colonic bacterial metabolism: an in vitro and in vivo study.,” Br. J. Nutr., vol. 74, no. 5, pp. 733–41, 1995.eng
dcterms.referencesFAO/WHO, “Guidelines for the evaluation of probiotics in food,” London World Heal. Organ. ON, Canada Food Agric. Organ., 2002.eng
dcterms.referencesS. Doron and S. L. Gorbach, “Probiotics: their role in the treatment and prevention of disease.,” Expert Rev. Anti. Infect. Ther., vol. 4, no. 2, pp. 261–275, 2006.eng
dcterms.referencesD. Y. Park et al., “Supplementation of Lactobacillus curvatus KY1032 in Diet-Induced Obese Mice Is Associated with Gut Microbial Changes and Reduction in Obesity,” PLoS One, vol. 8, no. 3, p. 59470, 2013.eng
dcterms.referencesD. Y. Park, Y. T. Ahn, C. S. Huh, R. A. Mcgregor, and M. S. Choi, “Dual probiotic strains suppress high fructoseinduced metabolic syndrome,” World J. Gastroenterol., vol. 19, no. 2, pp. 274–283, 2013.eng
dcterms.referencesA. Everard et al., “Cross-talk between Akkermansia muciniphila and intestinal epithelium controls dietinduced obesity.,” Proc. Natl. Acad. Sci. U. S. A., vol. 110, no. 22, pp. 9066–71, 2013.eng
dcterms.referencesP. G. Cano, A. Santacruz, A. Moya, and Y. Sanz, “Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity,” PLoS One, vol. 7, no. 7, p. e41079, 2012.eng
dcterms.referencesB. R. Goldin and S. L. Gorbach, “Alterations of the intestinal microflora by diet, oral antibiotics, and lactobacillus: Decreased production of free amines from aromatic nitro compounds, azo dyes, and glucuronides,” J. Natl. Cancer Inst., vol. 73, no. 3, pp. 689–695, 1984eng
dcterms.referencesB. R. Goldin and S. L. Gorbach, “The effect of milk and lactobacillus feeding on human intestinal bacterial enzyme activity,” Am. J. Clin. Nutr., vol. 39, no. 5, pp. 756–761, 1984.eng
dcterms.referencesI. A. Kirpich et al., “Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: a pilot study,” Alcohol, vol. 42, no. 8, pp. 675–682, 2008.eng
dcterms.referencesY. Wang et al., “Lactobacillus rhamnosus GG treatment potentiates intestinal hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury,” Am. J. Pathol., vol. 179, no. 6, pp. 2866–2875, 2011eng
dcterms.referencesM. Naruszewicz, M.-L. Johansson, D. Zapolska-Downar, and H. Bukowska, “Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers.,” Am. J. Clin. Nutr., vol. 76, no. 6, pp. 1249–55, 2002.eng
dcterms.referencesI. Kimura et al., “The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43.,” Nat. Commun., vol. 4, p. 1829, 2013.eng
dcterms.referencesK.-Y. Park, B. Kim, and C.-K. Hyun, “Lactobacillus rhamnosus GG improves glucose tolerance through alleviating ER stress and suppressing macrophage activation in db/db mice.,” J. Clin. Biochem. Nutr., vol. 56, no. 3, pp. 240–6, 2015.eng
dcterms.referencesK. A. Tappenden, D. M. Albin, A. L. Bartholome, and H. F. Mangian, “Glucagon-like peptide-2 and short-chain fatty acids: a new twist to an old story,” J Nutr, vol. 133, no. 11, pp. 3717–3720, 2003.eng
dcterms.referencesV. Marcil, E. Delvin, C. Garofalo, and E. Levy, “Butyrate impairs lipid transport by inhibiting microsomal triglyceride transfer protein in Caco-2 cells.,” J. Nutr., vol. 133, no. 7, pp. 2180–2183, 2003.eng
dcterms.referencesN. M. Delzenne, A. M. Neyrinck, and P. D. Cani, “Gut microbiota and metabolic disorders: How prebiotic can work?,” Br. J. Nutr., vol. 109 Suppl, pp. S81-5, 2013.eng
dcterms.referencesM. Galisteo, J. Duarte, and A. Zarzuelo, “Effects of dietary fibers on disturbances clustered in the metabolic syndrome,” Journal of Nutritional Biochemistry, vol. 19, no. 2. pp. 71–84, 2008.eng
dcterms.referencesE. Chambers et al., “Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults,” Gut, p. gutjnl, 2014.eng
dcterms.referencesG. Frost, D. Morrison, and T. Preston, “Compounds And Their Effects On Appetite Control And Insulin Sensitivity,” 2013.eng
dcterms.referencesY.-Y. Ma, L. Li, C. Yu, Z. Shen, L. Chen, and Y. Li, “Effects of probiotics on nonalcoholic fatty liver disease: a meta-analysis.,” World J. Gastroenterol., vol. 19, no. 40, pp. 6911–8, 2013eng
dcterms.referencesQ. Liu, Z. P. Duan, D. K. Ha, S. Bengmark, J. Kurtovic, and S. M. Riordan, “Synbiotic Modulation of Gut Flora: Effect on Minimal Hepatic Encephalopathy in Patients with Cirrhosis,” Hepatology, vol. 39, no. 5, pp. 1441– 1449, 2004.eng
dcterms.referencesZ. Asemi, A. Khorrami-Rad, and S. Alizadeh, “Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial,” Clin. Nutr., vol. 33, no. 2, pp. 198–203, 2014. -eng
dcterms.referencesY. Kadooka et al., “Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial,” Eur. J. Clin. Nutr., vol. 64, no. 10, pp. 636–643, 2010.eng
dcterms.referencesL. V. McFarland, “Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease,” Am. J. Gastroenterol., vol. 101, no. 4, pp. 812–822, 2006.eng
dcterms.references] M. Ito, K. Ohishi, Y. Yoshida, W. Yokoi, and H. Sawada, “Antioxidative effects of lactic acid bacteria on the colonic mucosa of iron-overloaded mice,” J. Agric. Food Chem., vol. 51, no. 15, pp. 4456–4460, 2003.eng
dcterms.referencesT. Kullisaar, J. Shepetova, and K. Zilmer, “An antioxidant probiotic reduces postprandial lipemia and oxidative stress,” Cent. Eur. J. Biol., vol. 6, no. 1, pp. 32–40, 2011.eng
dcterms.referencesH. Chung, J. Yu, I. Lee, and M. Liu, “Intestinal removal of free fatty acids from hosts by Lactobacilli for the treatment of obesity,” FEBS Open Bio, vol. 6, no. 1, pp. 64–76, 2016.eng
dcterms.referencesS. Thomas, D. Metzke, J. Schmitz, Y. Dörffel, and D. C. Baumgart, “Anti-inflammatory effects of Saccharomyces boulardii mediated by myeloid dendritic cells from patients with Crohn’s disease and ulcerative colitis.,” Am. J. Physiol. Gastrointest. Liver Physiol., vol. 301, no. 6, pp. G1083-92, 2011.eng
dcterms.referencesJ. P. Buts, N. Dekeyser, C. Stilmant, E. Delem, F. Smets, and E. Sokal, “Saccharomyces boulardii produces in rat small intestine a novel protein phosphatase that inhibits Escherichia coli endotoxin by dephosphorylation,” Pediatr. Res., vol. 60, no. 1, pp. 24–29, 2006.eng
dcterms.referencesM. Fujiya et al., “The Bacillus subtilis Quorum-Sensing Molecule CSF Contributes to Intestinal Homeostasis via OCTN2, a Host Cell Membrane Transporter,” Cell Host Microbe, vol. 1, no. 4, pp. 299–308, 2007.eng
dcterms.referencesA. Seth, F. Yan, D. Polk, and R. Rao, “Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruption by a PKC-and MAP kinase-dependent mechanism,” Am. J. Physiol. Liver Physiol., vol. 294, no. 4, pp. G1060–G1069, 2008.eng
dcterms.referencesE. O. Petrof et al., “Probiotics inhibit nuclear factor-kB and induce heat shock proteins in colonic epithelial cells through proteasome inhibition,” Gastroenterology, vol. 127, no. 5, pp. 1474–1487, 2004.eng
thesis.degree.disciplineFacultad de Ingenieríaes_CO
thesis.degree.levelDoctorado en Biocienciases_CO
thesis.degree.nameDoctor en Biocienciases_CO


Ficheros en el ítem

Thumbnail

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem

Attribution-NonCommercial-NoDerivatives 4.0 InternationalExcepto si se señala otra cosa, la licencia del ítem se describe como Attribution-NonCommercial-NoDerivatives 4.0 International