Transmissible microbial and metabolomic remodeling by soluble dietary fiber improves metabolic homeostasis

Baokun He, Kazunari Nohara, Nadim J Ajami, Ryan D Michalek, Xiangjun Tian, Matthew Wong, Susan H Losee-Olson, Joseph F Petrosino, Seung-Hee Yoo, Kazuhiro Shimomura, Zheng Chen, Baokun He, Kazunari Nohara, Nadim J Ajami, Ryan D Michalek, Xiangjun Tian, Matthew Wong, Susan H Losee-Olson, Joseph F Petrosino, Seung-Hee Yoo, Kazuhiro Shimomura, Zheng Chen

Abstract

Dietary fibers are increasingly appreciated as beneficial nutritional components. However, a requisite role of gut microbiota in fiber function and the overall impact of fibers on metabolomic flux remain unclear. We herein showed enhancing effects of a soluble resistant maltodextrin (RM) on glucose homeostasis in mouse metabolic disease models. Remarkably, fecal microbiota transplantation (FMT) caused pronounced and time-dependent improvement in glucose tolerance in RM recipient mice, indicating a causal relationship between microbial remodeling and metabolic efficacy. Microbial 16S sequencing revealed transmissible taxonomic changes correlated with improved metabolism, notably enrichment of probiotics and reduction of Alistipes and Bacteroides known to associate with high fat/protein diets. Metabolomic profiling further illustrated broad changes, including enrichment of phenylpropionates and decreases in key intermediates of glucose utilization, cholesterol biosynthesis and amino acid fermentation. These studies elucidate beneficial roles of RM-dependent microbial remodeling in metabolic homeostasis, and showcase prevalent health-promoting potentials of dietary fibers.

Figures

Figure 1. RM improves energy homeostasis and…
Figure 1. RM improves energy homeostasis and alters gut microbiota in db/db mice.
Fasting glucose (a), oral glucose tolerance test (OGTT) (b) and insulin tolerance test (ITT) (c) showed improvements in db/db mice after 8 weeks of RM treatment relative to Ctrl (n = 7-8). Area under curve (AUC) values are also shown for GTT and ITT. Values are presented as means ± SEM. *p < 0.05, **p < 0.01. For microbial sequencing analysis, alpha-diversity plots of gut microbiota (d) and heat maps of relative abundance of OTUs (as percentage of total microbiota) (e) are shown for db/db mice treated for 8 weeks with Ctrl or RM (n = 4-5). Level 2 (phylum) and 6 (genus) are shown. P values for diversity plots were calculated by 2-way ANOVA (repeated measure) to be Chao1 (P = 0.62) or Shannon (P = 0.59). See Supplementary Tables 1 and 2 for numerical values for heat maps in (e).
Figure 2. Fecal microbiota from donor db/db…
Figure 2. Fecal microbiota from donor db/db mice confers metabolic benefits of RM in recipient db/db mice.
(a) Alpha-diversity plots showed depletion of gut microbiota by antibiotics treatment in recipient mice prior to fecal microbiota transplantation. P values were calculated by 2-way ANOVA (repeat measure) to be Chao1 (P < 0.0001) or Shannon (P < 0.0001). Fasting glucose (b) and oral glucose tolerance test (OGTT) (c) in recipient db/db mice before (0 month) and after transplantation (1, 2 and 3 months) of fecal microbiota from donor mice (n = 6). Area under curve (AUC) values are also shown. Values are presented as means ± SEM. *p < 0.05, **p < 0.01.
Figure 3. Transmissible remodeling of gut microbiota…
Figure 3. Transmissible remodeling of gut microbiota in recipient db/db mice after fecal microbiota transplantation.
Alpha-diversity plots (a), heat maps of average (b) and time-resolved (c) abundance of phylum- and genus-level OTUs calculated as percentage of total microbiota in recipient db/db mice (n = 6) are shown. Values are presented as means ± SEM. P values for diversity plots were calculated by 2-way ANOVA (repeated measure) to be Chao1 (P = 0.014) or Shannon (P = 0.086). See Supplementary Tables 5 and 6 for numerical values for heat maps.
Figure 4. RM alters fecal metabolomic profiles…
Figure 4. RM alters fecal metabolomic profiles in donor and recipient db/db mice.
(a) Numbers of fecal metabolites affected, either up- or down-regulated (p < 0.05), by RM in donor and recipient mice (n = 6). Venn diagrams showing the overlaps between donor and recipient samples are also shown. (b) Principal component analysis of fecal metabolites affected by RM in donor and recipient mice (n = 6). (c) Hierarchical clustering heat map showing a predominant effect of RM on the relative abundance of the fecal metabolites such that RM donor and RM recipient mice (dRM and rRM), as well as Ctrl donor and Ctrl recipient (dCtrl and rCtrl), are clustered together (n = 6). Color bar values correspond to relative abundance measured in metabolomic analysis. Note that the color scheme is different from that for the heat maps showing microbial changes. (d) Random forest analysis showing a unique metabolomic signature between Ctrl and RM fecal samples, preserved in both donors and recipients, with a predictive accuracy of 96% in differentiating between the Ctrl and RM groups (n = 6).
Figure 5. RM alters metabolites involving in…
Figure 5. RM alters metabolites involving in glucose metabolism of gut microbiota in donor and recipient mice.
(a) Schematic of the glucose metabolism pathway. Metabolites decreased by RM treatment in the feces of donor or recipient db/db mice are highlighted in green whereas metabolites not detected or unchanged are marked in black or grey. (b) Graphs showing relative abundance of metabolites affected by RM treatment in the feces of donor or recipient db/db mice (n = 6). Values are presented as means ± SEM.
Figure 6. RM alters metabolites involving in…
Figure 6. RM alters metabolites involving in cholesterol metabolism of gut microbiota in donor and recipient mice.
(a) Schematic of the cholesterol metabolic pathway. Metabolites decreased by RM treatment in the feces of donor or recipient db/db mice are highlighted in green whereas metabolites not detected or unchanged are marked in black. (b) Graphs showing relative abundance of metabolites affected by RM treatment in the feces of donor or recipient db/db mice (n = 6). Values are presented as means ± SEM.

References

    1. Anderson J. W. et al. Health benefits of dietary fiber. Nutr Rev 67, 188–205, 10.1111/j.1753-4887.2009.00189.x (2009).
    1. Nicholson J. K. et al. Host-gut microbiota metabolic interactions. Science 336, 1262–1267, 10.1126/science.1223813 (2012).
    1. Flint H. J., Scott K. P., Louis P. & Duncan S. H. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol 9, 577–589, 10.1038/nrgastro.2012.156 (2012).
    1. Kishimoto Y., Oga H., Tagami H., Okuma K. & Gordon D. T. Suppressive effect of resistant maltodextrin on postprandial blood triacylglycerol elevation. Eur J Nutr 46, 133–138, 10.1007/s00394-007-0643-1 (2007).
    1. Fastinger N. D. et al. A novel resistant maltodextrin alters gastrointestinal tolerance factors, fecal characteristics, and fecal microbiota in healthy adult humans. J Am Coll Nutr 27, 356–366 (2008).
    1. Brown L., Rosner B., Willett W. W. & Sacks F. M. Cholesterol-lowering effects of dietary fiber: a meta-analysis. Am J Clin Nutr 69, 30–42 (1999).
    1. Chandalia M. et al. Beneficial effects of high dietary fiber intake in patients with type 2 diabetes mellitus. N Engl J Med 342, 1392–1398, 10.1056/NEJM200005113421903 (2000).
    1. De Vadder F. et al. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 156, 84–96, 10.1016/j.cell.2013.12.016 (2014).
    1. Cani P. D. et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr 90, 1236–1243, 10.3945/ajcn.2009.28095 (2009).
    1. Everard A. et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60, 2775–2786, 10.2337/db11-0227 (2011).
    1. Walsh C. J., Guinane C. M., O’Toole P. W. & Cotter P. D. Beneficial modulation of the gut microbiota. FEBS Lett, 10.1016/j.febslet.2014.03.035 (2014).
    1. Saulnier D. M., Spinler J. K., Gibson G. R. & Versalovic J. Mechanisms of probiosis and prebiosis: considerations for enhanced functional foods. Curr Opin Biotechnol 20, 135–141, 10.1016/j.copbio.2009.01.002 (2009).
    1. Tuohy K. M., Rouzaud G. C., Bruck W. M. & Gibson G. R. Modulation of the human gut microflora towards improved health using prebiotics--assessment of efficacy. Curr Pharm Des 11, 75–90 (2005).
    1. Scott K. P., Duncan S. H., Louis P. & Flint H. J. Nutritional influences on the gut microbiota and the consequences for gastrointestinal health. Biochem Soc Trans 39, 1073–1078, 10.1042/BST0391073 (2011).
    1. Koropatkin N. M., Cameron E. A. & Martens E. C. How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol 10, 323–335, 10.1038/nrmicro2746 (2012).
    1. Shen J., Obin M. S. & Zhao L. The gut microbiota, obesity and insulin resistance. Mol Aspects Med 34, 39–58, 10.1016/j.mam.2012.11.001 (2013).
    1. Ridaura V. K. et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341, 1241214, 10.1126/science.1241214 (2013).
    1. Macfarlane S. & Macfarlane G. T. Regulation of short-chain fatty acid production. Proc Nutr Soc 62, 67–72, 10.1079/PNS2002207 (2003).
    1. Donohoe D. R. et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab 13, 517–526, 10.1016/j.cmet.2011.02.018 (2011).
    1. Maslowski K. M. et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282–1286, 10.1038/nature08530 (2009).
    1. Trompette A. et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20, 159–166, 10.1038/nm.3444 (2014).
    1. Frost G. et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 5, 3611, 10.1038/ncomms4611 (2014).
    1. Russell W. R., Hoyles L., Flint H. J. & Dumas M. E. Colonic bacterial metabolites and human health. Curr Opin Microbiol 16, 246–254, 10.1016/j.mib.2013.07.002 (2013).
    1. Jacobs D. M., Gaudier E., van Duynhoven J. & Vaughan E. E. Non-digestible food ingredients, colonic microbiota and the impact on gut health and immunity: a role for metabolomics. Curr Drug Metab 10, 41–54 (2009).
    1. Polakof S. et al. Resistant starch intake partly restores metabolic and inflammatory alterations in the liver of high-fat-diet-fed rats. J Nutr Biochem 24, 1920–1930, 10.1016/j.jnutbio.2013.05.008 (2013).
    1. Martin F. P. et al. Dietary modulation of gut functional ecology studied by fecal metabonomics. J Proteome Res 9, 5284–5295, 10.1021/pr100554m (2010).
    1. Baer D. J. et al. The metabolizable energy of dietary resistant maltodextrin is variable and alters fecal microbiota composition in adult men. J Nutr 144, 1023–1029, 10.3945/jn.113.185298 (2014).
    1. Delzenne N. M., Cani P. D. & Neyrinck A. M. Modulation of glucagon-like peptide 1 and energy metabolism by inulin and oligofructose: experimental data. J Nutr 137, 2547S–2551S (2007).
    1. Cotillard A. et al. Dietary intervention impact on gut microbial gene richness. Nature 500, 585–588, 10.1038/nature12480 (2013).
    1. Le Chatelier E. et al. Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546, 10.1038/nature12506 (2013).
    1. Larsen N. et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 5, e9085, 10.1371/journal.pone.0009085 (2010).
    1. David L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563, 10.1038/nature12820 (2014).
    1. Willing B. P., Vacharaksa A., Croxen M., Thanachayanont T. & Finlay B. B. Altering host resistance to infections through microbial transplantation. PLoS One 6, e26988, 10.1371/journal.pone.0026988 (2011).
    1. Ghosh S. et al. Colonic microbiota alters host susceptibility to infectious colitis by modulating inflammation, redox status, and ion transporter gene expression. Am J Physiol Gastrointest Liver Physiol 301, G39–49, 10.1152/ajpgi.00509.2010 (2011).
    1. Smith E. A. & Macfarlane G. T. Formation of Phenolic and Indolic Compounds by Anaerobic Bacteria in the Human Large Intestine. Microbial ecology 33, 180–188 (1997).
    1. Russell W. R. et al. Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Molecular nutrition & food research 57, 523–535, 10.1002/mnfr.201200594 (2013).
    1. Hashimoto S. et al. Effects of beta-lactam antibiotics on intestinal microflora and bile acid metabolism in rats. Lipids 31, 601–609 (1996).
    1. Kishimoto Y., Wakabayashi S. & Takeda H. Hypocholesterolemic effect of dietary fiber: relation to intestinal fermentation and bile acid excretion. J Nutr Sci Vitaminol (Tokyo) 41, 151–161 (1995).
    1. Mayer E. A., Savidge T. & Shulman R. J. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology 146, 1500–1512, 10.1053/j.gastro.2014.02.037 (2014).
    1. Perez-Cobas A. E. et al. Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut 62, 1591–1601, 10.1136/gutjnl-2012-303184 (2013).
    1. Qin J. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60, 10.1038/nature11450 (2012).
    1. Zhang X. et al. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS One 8, e71108, 10.1371/journal.pone.0071108 (2013).
    1. Wu X. et al. Molecular characterisation of the faecal microbiota in patients with type II diabetes. Curr Microbiol 61, 69–78, 10.1007/s00284-010-9582-9 (2010).
    1. Schwiertz A. et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 18, 190–195, 10.1038/oby.2009.167 (2010).
    1. Turnbaugh P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484, 10.1038/nature07540 (2009).
    1. Karlsson F. H. et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99–103, 10.1038/nature12198 (2013).
    1. Holmes E., Li J. V., Marchesi J. R. & Nicholson J. K. Gut microbiota composition and activity in relation to host metabolic phenotype and disease risk. Cell Metab 16, 559–564, 10.1016/j.cmet.2012.10.007 (2012).
    1. Tremaroli V. & Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature 489, 242–249, 10.1038/nature11552 (2012).
    1. McHardy I. H. et al. Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. Microbiome 1, 17, 10.1186/2049-2618-1-17 (2013).
    1. Jenner A. M., Rafter J. & Halliwell B. Human fecal water content of phenolics: the extent of colonic exposure to aromatic compounds. Free radical biology & medicine 38, 763–772, 10.1016/j.freeradbiomed.2004.11.020 (2005).
    1. Metges C. C. & Petzke K. J. Utilization of essential amino acids synthesized in the intestinal microbiota of monogastric mammals. The British journal of nutrition 94, 621–622 (2005).
    1. Ratia K. et al. Discovery of selective inhibitors of the Clostridium difficile dehydroquinate dehydratase. PLoS One 9, e89356, 10.1371/journal.pone.0089356 (2014).
    1. Zhou J., Bowler L. D. & Spratt B. G. Interspecies recombination, and phylogenetic distortions, within the glutamine synthetase and shikimate dehydrogenase genes of Neisseria meningitidis and commensal Neisseria species. Molecular microbiology 23, 799–812 (1997).
    1. Flickinger E. A. et al. Glucose-based oligosaccharides exhibit different in vitro fermentation patterns and affect in vivo apparent nutrient digestibility and microbial populations in dogs. J Nutr 130, 1267–1273 (2000).
    1. Backhed F., Ley R. E., Sonnenburg J. L., Peterson D. A. & Gordon J. I. Host-bacterial mutualism in the human intestine. Science 307, 1915–1920, 10.1126/science.1104816 (2005).
    1. Caporaso J. G. et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. The ISME journal 6, 1621–1624, 10.1038/ismej.2012.8 (2012).
    1. Edgar R. C. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nature methods 10, 996–998, 10.1038/nmeth.2604 (2013).
    1. Evans A. M., DeHaven C. D., Barrett T., Mitchell M. & Milgram E. Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems. Anal Chem 81, 6656–6667, 10.1021/ac901536h (2009).
    1. Dehaven C. D., Evans A. M., Dai H. & Lawton K. A. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. J Cheminform 2, 9, 10.1186/1758-2946-2-9 (2010).
    1. Touw W. G. et al. Data mining in the Life Sciences with Random Forest: a walk in the park or lost in the jungle? Briefings in bioinformatics 14, 315–326, 10.1093/bib/bbs034 (2013).

Source: PubMed

3
Abonneren