Minor Changes in the Composition and Function of the Gut Microbiota During a 12-Week Whole Grain Wheat or Refined Wheat Intervention Correlate with Liver Fat in Overweight and Obese Adults

Mara P H van Trijp, Sophie Schutte, Diederik Esser, Suzan Wopereis, Femke P M Hoevenaars, Guido J E J Hooiveld, Lydia A Afman, Mara P H van Trijp, Sophie Schutte, Diederik Esser, Suzan Wopereis, Femke P M Hoevenaars, Guido J E J Hooiveld, Lydia A Afman

Abstract

Background: Whole grain wheat (WGW) products are advocated as a healthy choice when compared with refined wheat (RW). One proposed mechanism for these health benefits is via the microbiota, because WGW contains multiple fibers. WGW consumption has been proposed to ameliorate nonalcoholic fatty liver disease, in which microbiota might play a role.

Objectives: We investigated the effect of WGW compared with RW intervention on the fecal microbiota composition and functionality, and correlated intervention-induced changes in bacteria with changes in liver health parameters in adults with overweight or obesity.

Methods: We used data of a 12-wk double-blind, randomized, controlled, parallel trial to examine the effects of a WGW (98 g/d) or RW (98 g/d) intervention on the secondary outcomes fecal microbiota composition, predicted microbiota functionality, and stool consistency in 37 women and men (aged 45-70 y, BMI 25-35 kg/m2). The changes in microbiota composition, measured using 16S ribosomal RNA gene sequencing, after a 12-wk intervention were analyzed with nonparametric tests, and correlated with changes in liver fat and circulating concentrations of liver enzymes including alanine transaminase, aspartate transaminase, γ-glutamyltransferase, and serum amyloid A.

Results: The WGW intervention increased the mean (± SD) relative abundances of Ruminococcaceae_UCG-014 (baseline: 2.2 ± 4.6%, differential change over time (Δ) 0.51 ± 4.2%), Ruminiclostridium_9 (baseline: 0.065 ± 0.11%, Δ 0.054 ± 0.14%), and Ruminococcaceae_NK4A214_group (baseline: 0.37 ± 0.56%, Δ 0.17 ± 0.83%), and also the predicted pathway acetyl-CoA fermentation to butyrate II (baseline: 0.23 ± 0.062%, Δ 0.035 ± 0.059%), compared with the RW intervention (P values <0.05). A change in Ruminococcaceae_NK4A214_group was positively correlated with the change in liver fat, in both the WGW (ρ = 0.54; P = 0.026) and RW (ρ = 0.67; P = 0.024) groups.

Conclusions: In middle-aged overweight and obese adults, a 12-wk WGW intervention increased the relative abundance of a number of bacterial taxa from the family Ruminococcaceae and increased predicted fermentation pathways when compared with an RW intervention. Potential protective health effects of replacement of RW by WGW on metabolic organs, such as the liver, via modulation of the microbiota, deserve further investigation.This trial was registered at clinicaltrials.gov as NCT02385149.

Keywords: fatty liver; fermentation; gut microbiota; human; overweight/obesity; refined wheat; whole grain wheat.

© The Author(s) 2020. Published by Oxford University Press on behalf of the American Society for Nutrition.

Figures

FIGURE 1
FIGURE 1
Significantly different fecal bacterial taxa and bacterial pathways at baseline and after 12-wk of an RW or WGW intervention in middle-aged overweight and obese adults. The flow diagram shows the number of bacterial taxa at the genus level and the predicted pathways for which the change in relative abundance was significantly different among interventions. RW, refined wheat; WGW, whole grain wheat.
FIGURE 2
FIGURE 2
The effect of 12 wk of an RW or WGW intervention on the fecal microbiota diversity in middle-aged overweight and obese adults. (A) PCoA plot using weighted UniFrac dissimilarity to visualize the overall microbiota community variation. Color and shape highlight the intervention groups before or after the intervention. The lines connect the within-person samples over time; 95% CIs are plotted. (B) Microbial diversity as assessed using Faith phylogenetic diversity at baseline and after the 12-wk intervention (end). Individual paired samples are connected by a line. The width of the colored shapes indicates the sample density, the squared shape inside indicates the group mean. PCoA, principal coordinate analysis; RW, refined wheat; WGW, whole grain wheat.
FIGURE 3
FIGURE 3
Fecal bacterial taxa at the genus level at baseline and after the 12-wk RW or WGW intervention (end) that were found to be significantly different between the groups in middle-aged overweight and obese adults. The relative abundances of (A) Ruminococcaceae_NK4A214_group; (B) Ruminococcaceae_UCG-014; (C) Lachnospiraceae_UCG-008; and (D) Ruminiclostridium_9 are shown. Data are presented as group mean (the squared shape), n = 16 (RW) or n = 21 (WGW), and the width of the colored shapes indicates the sample density. Individual paired samples are connected by a line. RW, refined wheat; WGW, whole grain wheat.
FIGURE 4
FIGURE 4
Fecal bacterial taxa at the genus level that were found to be significantly correlated with IHTGs at baseline in middle-aged overweight and obese adults. The relative abundances of (A) Roseburia; (B) Ruminococcus_2; (C) Faecalibacterium; (D) Ruminococcaceae_UCG-010; (E) Ruminococcaceae_UCG-005; and (F) Akkermansia are shown, fitted with a linear regression model with a 95% CI. Noncorrected P values are shown. Data of n = 35 participants are shown, who had both microbiota and IHTG data at baseline available. IHTG, intrahepatic triglyceride.
FIGURE 5
FIGURE 5
Predicted fecal microbial pathway relative abundance at baseline and after 12 wk of RW or WGW intervention that were found to be significantly different between the groups in middle-aged overweight and obese adults. The relative abundances of the predicted pathways (A) superpathway of hexitol degradation; (B) pantothenate and coenzyme A biosynthesis; (C) acetyl-CoA fermentation to butyrate II; (D) pyruvate fermentation to acetone; (E) aromatic biogenic amine degradation; and (F) l-alanine biosynthesis are shown. Data are presented as group mean (the squared shape), n = 16 (RW) or n = 21 (WGW), and the width of the colored shapes indicates the sample density. Individual paired samples are connected by a line. RW, refined wheat; WGW, whole grain wheat.

References

    1. de Munter JS, Hu FB, Spiegelman D, Franz M, van Dam RM. Whole grain, bran, and germ intake and risk of type 2 diabetes: a prospective cohort study and systematic review. PLoS Med. 2007;4(8):e261.
    1. Truswell A. Cereal grains and coronary heart disease. Eur J Clin Nutr. 2002;56(1):1.
    1. Sahyoun NR, Jacques PF, Zhang XL, Juan W, McKeown NM. Whole-grain intake is inversely associated with the metabolic syndrome and mortality in older adults. Am J Clin Nutr. 2006;83(1):124–31.
    1. Ross AB, Godin J-P, Minehira K, Kirwan JP. Increasing whole grain intake as part of prevention and treatment of nonalcoholic fatty liver disease. Int J Endocrinol. 2013;2013:585876.
    1. Bartłomiej S, Justyna R-K, Ewa N. Bioactive compounds in cereal grains—occurrence, structure, technological significance and nutritional benefits—a review. Food Sci Technol Int. 2012;18(6):559–68.
    1. McKevith B. Nutritional aspects of cereals. Nutr Bull. 2004;29(2):111–42.
    1. Schutte S, Esser D, Hoevenaars FP, Hooiveld GJ, Priebe MG, Vonk RJ, Wopereis S, Afman LA. A 12-wk whole-grain wheat intervention protects against hepatic fat: the Graandioos study, a randomized trial in overweight subjects. Am J Clin Nutr. 2018;108(6):1264–74.
    1. Kirwan JP, Malin SK, Scelsi AR, Kullman EL, Navaneethan SD, Pagadala MR, Haus JM, Filion J, Godin JP, Kochhar S et al. A whole-grain diet reduces cardiovascular risk factors in overweight and obese adults: a randomized controlled trial. J Nutr. 2016;146(11):2244–51.
    1. Tighe P, Duthie G, Vaughan N, Brittenden J, Simpson WG, Duthie S, Mutch W, Wahle K, Horgan G, Thies F. Effect of increased consumption of whole-grain foods on blood pressure and other cardiovascular risk markers in healthy middle-aged persons: a randomized controlled trial. Am J Clin Nutr. 2010;92(4):733–40.
    1. Hollænder PL, Ross AB, Kristensen M. Whole-grain and blood lipid changes in apparently healthy adults: a systematic review and meta-analysis of randomized controlled studies. Am J Clin Nutr. 2015;102(3):556–72.
    1. Fardet A. New hypotheses for the health-protective mechanisms of whole-grain cereals: what is beyond fibre? Nutr Res Rev. 2010;23(1):65–134.
    1. Makki K, Deehan EC, Walter J, Bäckhed F. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe. 2018;23(6):705–15.
    1. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut microbes. 2017;8(2):172–84.
    1. Kortman GA, Dutilh BE, Maathuis AJ, Engelke UF, Boekhorst J, Keegan KP, Nielsen FG, Betley J, Weir JC, Kingsbury Z. Microbial metabolism shifts towards an adverse profile with supplementary iron in the TIM-2 in vitro model of the human colon. Front Microbiol. 2016;6:1481.
    1. Fehlbaum S, Prudence K, Kieboom J, Heerikhuisen M, van den Broek T, Schuren F, Steinert R, Raederstorff D. In vitro fermentation of selected prebiotics and their effects on the composition and activity of the adult gut microbiota. IJMS. 2018;19(10):3097.
    1. Jefferson A, Adolphus K. The effects of intact cereal grain fibers, including wheat bran on the gut microbiota composition of healthy adults: a systematic review. Front Nutr. 2019;6(33).
    1. Christensen EG, Licht TR, Kristensen M, Bahl MI. Bifidogenic effect of whole-grain wheat during a 12-week energy-restricted dietary intervention in postmenopausal women. Eur J Clin Nutr. 2013;67(12):1316–21.
    1. Costabile A, Klinder A, Fava F, Napolitano A, Fogliano V, Leonard C, Gibson GR, Tuohy KM. Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: a double-blind, placebo-controlled, crossover study. Br J Nutr. 2008;99(1):110–20.
    1. Vitaglione P, Mennella I, Ferracane R, Rivellese AA, Giacco R, Ercolini D, Gibbons SM, La Storia A, Gilbert JA, Jonnalagadda S et al. Whole-grain wheat consumption reduces inflammation in a randomized controlled trial on overweight and obese subjects with unhealthy dietary and lifestyle behaviors: role of polyphenols bound to cereal dietary fiber. Am J Clin Nutr. 2015;101(2):251–61.
    1. Vanegas SM, Meydani M, Barnett JB, Goldin B, Kane A, Rasmussen H, Brown C, Vangay P, Knights D, Jonnalagadda S. Substituting whole grains for refined grains in a 6-wk randomized trial has a modest effect on gut microbiota and immune and inflammatory markers of healthy adults. Am J Clin Nutr. 2017;105(3):635–50.
    1. Lappi J, Salojärvi J, Kolehmainen M, Mykkänen H, Poutanen K, de Vos WM, Salonen A. Intake of whole-grain and fiber-rich rye bread versus refined wheat bread does not differentiate intestinal microbiota composition in Finnish adults with metabolic syndrome. J Nutr. 2013;143(5):648–55.
    1. Ampatzoglou A, Atwal KK, Maidens CM, Williams CL, Ross AB, Thielecke F, Jonnalagadda SS, Kennedy OB, Yaqoob P. Increased whole grain consumption does not affect blood biochemistry, body composition, or gut microbiology in healthy, low-habitual whole grain consumers. J Nutr. 2015;145(2):215–21.
    1. Cooper DN, Kable ME, Marco ML, De Leon A, Rust B, Baker JE, Horn W, Burnett D, Keim NL. The effects of moderate whole grain consumption on fasting glucose and lipids, gastrointestinal symptoms, and microbiota. Nutrients. 2017;9(2):173.
    1. Bird AR, Vuaran MS, King RA, Noakes M, Keogh J, Morell MK, Topping DL. Wholegrain foods made from a novel high-amylose barley variety (Himalaya 292) improve indices of bowel health in human subjects. Br J Nutr. 2008;99(5):1032–40.
    1. Slavin J. Why whole grains are protective: biological mechanisms. Proc Nutr Soc. 2003;62(1):129–34.
    1. Boets E, Gomand SV, Deroover L, Preston T, Vermeulen K, Preter V, Hamer HM, den Mooter G, Vuyst L, Courtin CM. Systemic availability and metabolism of colonic‐derived short‐chain fatty acids in healthy subjects: a stable isotope study. J Physiol. 2017;595(2):541–55.
    1. Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. 2015;11(10):577.
    1. den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud D-J. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes. 2015;64(7):2398–408.
    1. Mollica MP, Raso GM, Cavaliere G, Trinchese G, De Filippo C, Aceto S, Prisco M, Pirozzi C, Di Guida F, Lama A. Butyrate regulates liver mitochondrial function, efficiency, and dynamic, in insulin resistant obese mice. Diabetes. 2017;66(5):1405–18.
    1. Kobyliak N, Abenavoli L, Mykhalchyshyn G, Kononenko L, Boccuto L, Kyriienko D, Dynnyk O. A multi-strain probiotic reduces the fatty liver index, cytokines and aminotransferase levels in NAFLD patients: evidence from a randomized clinical trial. J Gastrointestin Liver Dis. 2018;27(1):41–9.
    1. Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, Mastrojeni S, Malaguarnera G, Mistretta A, Li Volti G et al. Bifidobacterium longum with fructo-oligosaccharides in patients with non alcoholic steatohepatitis. Dig Dis Sci. 2012;57(2):545–53.
    1. Manzhalii E, Virchenko O, Falalyeyeva T, Beregova T, Stremmel W. Treatment efficacy of a probiotic preparation for non-alcoholic steatohepatitis: a pilot trial. J Dig Dis. 2017;18(12):698–703.
    1. Aller R, De Luis DA, Izaola O, Conde R, Gonzalez Sagrado M, Primo D, De La Fuente B, Gonzalez J. Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver disease patients: a double blind randomized clinical trial. Eur Rev Med Pharmacol Sci. 2011;15(9):1090–5.
    1. Hoevenaars FPM, Esser D, Schutte S, Priebe MG, Vonk RJ, van den Brink WJ, van der Kamp JW, Stroeve JHM, Afman LA, Wopereis S. Whole grain wheat consumption affects postprandial inflammatory response in a randomized controlled trial in overweight and obese adults with mild hypercholesterolemia in the Graandioos study. J Nutr. 2019;149(12):2133–44.
    1. Ramiro-Garcia J, Hermes GD, Giatsis C, Sipkema D, Zoetendal EG, Schaap PJ, Smidt H. NG-Tax, a highly accurate and validated pipeline for analysis of 16S rRNA amplicons from complex biomes. F1000Res. 2018;5:1791.
    1. Poncheewin W, Hermes GDA, van Dam JCJ, Koehorst JJ, Smidt H, Schaap PJ. NG-Tax 2.0: a semantic framework for high-throughput amplicon analysis. Front Genet. 2019;10:1366.
    1. Team RC . R: A language and environment for statistical computing. Vienna (Austria): R Foundation for Statistical Computing; 2007.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8(4):e61217.
    1. Weiss S, Xu ZZ, Peddada S, Amir A, Bittinger K, Gonzalez A, Lozupone C, Zaneveld JR, Vázquez-Baeza Y, Birmingham A et al. Normalization and microbial differential abundance strategies depend upon data characteristics. Microbiome. 2017;5(1):27.
    1. Oksanen J, Blanchet F, Friendly M, Kindt R, Legendre P, McGlinn D, Minchin P, O'Hara R, Simpson G, Solymos P. vegan: Community Ecology Package. R package version 2.5-2. R Foundation; 2018.
    1. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. 2013;31:814.
    1. Salonen A, Lahti L, Salojärvi J, Holtrop G, Korpela K, Duncan SH, Date P, Farquharson F, Johnstone AM, Lobley GE et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014;8:2218.
    1. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P, Bergerat A et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5:220.
    1. Vos P, Garrity G, Jones D, Krieg NR, Ludwig W, Rainey FA, Schleifer K-H, Whitman WB. Bergey's manual of systematic bacteriology. Vol. 3. The Firmicutes: Springer Science & Business Media; 2011.
    1. Robert C, Bernalier-Donadille A. The cellulolytic microflora of the human colon: evidence of microcrystalline cellulose-degrading bacteria in methane-excreting subjects. FEMS Microbiol Ecol. 2003;46(1):81–9.
    1. Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3(4):289–306.
    1. Vuholm S, Nielsen DS, Iversen KN, Suhr J, Westermann P, Krych L, Andersen JR, Kristensen M. Whole-grain rye and wheat affect some markers of gut health without altering the fecal microbiota in healthy overweight adults: a 6-week randomized trial. J Nutr. 2017;147(11):2067–75.
    1. Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, Greenwood R, Sikaroodi M, Lam V, Crotty P et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2013;11(7):868–75.e3.
    1. Jiang W, Wu N, Wang X, Chi Y, Zhang Y, Qiu X, Hu Y, Li J, Liu Y. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep. 2015;5(1):8096.
    1. Iino C, Endo T, Mikami K, Hasegawa T, Kimura M, Sawada N, Nakaji S, Fukuda S. Significant decrease in Faecalibacterium among gut microbiota in nonalcoholic fatty liver disease: a large BMI- and sex-matched population study. Hepatol Int. 2019;13(6):748–56.
    1. Korem T, Zeevi D, Zmora N, Weissbrod O, Bar N, Lotan-Pompan M, Avnit-Sagi T, Kosower N, Malka G, Rein M. Bread affects clinical parameters and induces gut microbiome-associated personal glycemic responses. Cell Metab. 2017;25(6):1243.
    1. Vandeputte D, Falony G, Vieira-Silva S, Wang J, Sailer M, Theis S, Verbeke K, Raes J. Prebiotic inulin-type fructans induce specific changes in the human gut microbiota. Gut. 2017;66(11):1968–74.
    1. Markowiak P, Śliżewska K. Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients. 2017;9(9):1021.
    1. Vital M, Karch A, Pieper DH. Colonic butyrate-producing communities in humans: an overview using omics data. mSystems. 2017;2(6):e00130–17.
    1. Muñoz-Tamayo R, Laroche B, Walter E, Doré J, Duncan SH, Flint HJ, Leclerc M. Kinetic modelling of lactate utilization and butyrate production by key human colonic bacterial species. FEMS Microbiol Ecol. 2011;76(3):615–24.
    1. Han F, Wang Y, Han Y, Zhao J, Han F, Song G, Jiang P, Miao H. Effects of whole-grain rice and wheat on composition of gut microbiota and short-chain fatty acids in rats. J Agric Food Chem. 2018;66(25):6326–35.
    1. Rose DJ. Impact of whole grains on the gut microbiota: the next frontier for oats? Br J Nutr. 2014;112(S2):S44–S9.
    1. Andersson AA, Andersson R, Piironen V, Lampi A-M, Nyström L, Boros D, Fraś A, Gebruers K, Courtin CM, Delcour JA. Contents of dietary fibre components and their relation to associated bioactive components in whole grain wheat samples from the HEALTHGRAIN diversity screen. Food Chem. 2013;136(3-4):1243–8.
    1. Andersson R, Fransson G, Tietjen M, Aman P. Content and molecular-weight distribution of dietary fiber components in whole-grain rye flour and bread. J Agric Food Chem. 2009;57(5):2004–8.
    1. Eriksen AK, Brunius C, Mazidi M, Hellström PM, Risérus U, Iversen KN, Fristedt R, Sun L, Huang Y, Nørskov NP et al. Effects of whole-grain wheat, rye, and lignan supplementation on cardiometabolic risk factors in men with metabolic syndrome: a randomized crossover trial. Am J Clin Nutr. 2020;111(4):864–76.
    1. Bach Knudsen KE, Lærke HN, Hedemann MS, Nielsen TS, Ingerslev AK, Gundelund Nielsen DS, Theil PK, Purup S, Hald S, Schioldan AG et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients. 2018;10(10):1499.
    1. Louis P, Flint HJ. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett. 2009;294(1):1–8.
    1. Wang B, Jiang X, Cao M, Ge J, Bao Q, Tang L, Chen Y, Li L. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non-alcoholic fatty liver disease. Sci Rep. 2016;6:32002.
    1. Del Chierico F, Nobili V, Vernocchi P, Russo A, De Stefanis C, Gnani D, Furlanello C, Zandonà A, Paci P, Capuani G et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology. 2017;65(2):451–64.
    1. Nistal E, Sáenz de Miera LE, Ballesteros Pomar M, Sánchez-Campos S, García-Mediavilla MV, Álvarez-Cuenllas B, Linares P, Olcoz JL, Arias-Loste MT, García-Lobo JM et al. An altered fecal microbiota profile in patients with non-alcoholic fatty liver disease (NAFLD) associated with obesity. Rev Esp Enferm Dig. 2019;111(4):275–82.

Source: PubMed

3
購読する