Mediterranean diet intervention in overweight and obese subjects lowers plasma cholesterol and causes changes in the gut microbiome and metabolome independently of energy intake

Victoria Meslier, Manolo Laiola, Henrik Munch Roager, Francesca De Filippis, Hugo Roume, Benoit Quinquis, Rosalba Giacco, Ilario Mennella, Rosalia Ferracane, Nicolas Pons, Edoardo Pasolli, Angela Rivellese, Lars Ove Dragsted, Paola Vitaglione, Stanislav Dusko Ehrlich, Danilo Ercolini, Victoria Meslier, Manolo Laiola, Henrik Munch Roager, Francesca De Filippis, Hugo Roume, Benoit Quinquis, Rosalba Giacco, Ilario Mennella, Rosalia Ferracane, Nicolas Pons, Edoardo Pasolli, Angela Rivellese, Lars Ove Dragsted, Paola Vitaglione, Stanislav Dusko Ehrlich, Danilo Ercolini

Abstract

Objectives: This study aimed to explore the effects of an isocaloric Mediterranean diet (MD) intervention on metabolic health, gut microbiome and systemic metabolome in subjects with lifestyle risk factors for metabolic disease.

Design: Eighty-two healthy overweight and obese subjects with a habitually low intake of fruit and vegetables and a sedentary lifestyle participated in a parallel 8-week randomised controlled trial. Forty-three participants consumed an MD tailored to their habitual energy intakes (MedD), and 39 maintained their regular diets (ConD). Dietary adherence, metabolic parameters, gut microbiome and systemic metabolome were monitored over the study period.

Results: Increased MD adherence in the MedD group successfully reprogrammed subjects' intake of fibre and animal proteins. Compliance was confirmed by lowered levels of carnitine in plasma and urine. Significant reductions in plasma cholesterol (primary outcome) and faecal bile acids occurred in the MedD compared with the ConD group. Shotgun metagenomics showed gut microbiome changes that reflected individual MD adherence and increase in gene richness in participants who reduced systemic inflammation over the intervention. The MD intervention led to increased levels of the fibre-degrading Faecalibacterium prausnitzii and of genes for microbial carbohydrate degradation linked to butyrate metabolism. The dietary changes in the MedD group led to increased urinary urolithins, faecal bile acid degradation and insulin sensitivity that co-varied with specific microbial taxa.

Conclusion: Switching subjects to an MD while maintaining their energy intake reduced their blood cholesterol and caused multiple changes in their microbiome and metabolome that are relevant in future strategies for the improvement of metabolic health.

Keywords: diet; intestinal microbiology; nutrition.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Adherence to the Mediterranean diet (MD) and changes in dietary and metabolic variables. (A) Box plots showing MD index score for controls (ConD) or treated subjects (MedD) during the intervention, the significance was tested by applying the post hoc Friedman-Nemenyi test for pairwise test of multiple comparisons within each group. (B) Percentage changes in dietary and metabolic variables are represented as spider chart. Changes in levels of dietary components consumption including (C) dietary fibre, (D) vegetable proteins/animal proteins ratio, (E) saturated to polyunsaturated fats ratio. Reduction in serum and urinary markers such as (F) plasma carnitine, (G) urinary carnitine and (H) total cholesterol. The significance was tested by applying unpaired Wilcoxon rank-sum tests for variation at the specific time point compared with baseline in MedD versus ConD. Orange boxes refer to controls and green boxes to Mediterranean subjects, respectively. Baseline, 0 weeks; 4w, 4 weeks; 8w, 8 weeks of nutritional intervention (*p

Figure 2

Mediterranean diet changes the intestinal…

Figure 2

Mediterranean diet changes the intestinal and systemic metabolome. Partial least squares discriminant analysis…

Figure 2
Mediterranean diet changes the intestinal and systemic metabolome. Partial least squares discriminant analysis plots based on molecular features detected in (A) faeces and (B) urine. Subjects belonging to different categories were coloured according to diet and time points: MedD subjects at baseline (light green), after 4 weeks (green) and 8 weeks of intervention (dark green). ConD subjects at baseline (light orange), after 4 weeks (orange) and 8 weeks (dark orange) of intervention. The loading plots display vectors that contributed the most to variability of individual dataset; variables explaining the variance between the groups in (C) faecal and (D) urine metabolome are reported as bar plots.

Figure 3

Microbial diversity richness anticorrelates with…

Figure 3

Microbial diversity richness anticorrelates with inflammation. (A) Spearman’s correlation between variation of gut…

Figure 3
Microbial diversity richness anticorrelates with inflammation. (A) Spearman’s correlation between variation of gut microbial gene richness and individual inflammatory status (serum hs-CRP) variation at the end of trial; n observation=62. (B) Violin plot showing differences in serum hs-CRP variation between subjects increasing (n=25, yellow) compared with subjects decreasing (n=37, light blue) gene richness at the end of trial. Statistical differences between groups were determined using unpaired Wilcoxon rank-sum tests. hs-CRP, high sensitivity C reactive protein.

Figure 4

Mediterranean diet (MD) affects gut…

Figure 4

Mediterranean diet (MD) affects gut microbiome composition. (A) Total delta MD index changes…

Figure 4
Mediterranean diet (MD) affects gut microbiome composition. (A) Total delta MD index changes over the 4w-baseline period. Left: histograms of delta (4w-baseline) MD index (n=62). Right: linear regressions of microbiome similarity compared with delta 4w-baseline MD index. Microbiome similarity was estimated by Spearman correlations between microbial composition at 4 weeks and baseline within each individual. (B) Total MD index fractional changes (FCs) (4w-baseline)/baseline, used as proxy to measure the effort of adherence (n=62). Left: distribution of individuals relative to MD index FC. Right: linear regressions of microbiome similarity and FC. Spearman correlations (rho and p values) are reported, excluding outliers, for ConD (n=31) and MedD (n=26) groups, respectively. 4w, 4 weeks.

Figure 5

Faecal BA concentrations over the…

Figure 5

Faecal BA concentrations over the nutritional intervention. Parallel coordinate plot showing variations of…

Figure 5
Faecal BA concentrations over the nutritional intervention. Parallel coordinate plot showing variations of faecal (A) total, (B) primary and (C) secondary BA concentrations within the MedD group during the intervention. The red triangles indicate mean values, the lines connecting dots are used to indicate the same sample at each time point. The significance was tested by applying the post hoc Friedman-Nemenyi test for pairwise test of multiple comparisons within each group. (D) In the box plot the relative abundances of Bilophila wadsworthia are compared considering subjects falling in the highest quartile (n=16, green) and in the lowest quartile of reduction (n=16, blue) of secondary to primary BAs ratio after 4 weeks of treatment. Baseline, 0 weeks; 4w, 4 weeks; 8w, 8 weeks of nutritional intervention. H, highest quartile of reduction; L, lowest quartile of reduction; BAs, bile acids.

Figure 6

MD intervention determines a reduction…

Figure 6

MD intervention determines a reduction of faecal branched-chain fatty acid (BCFA) concentrations and…

Figure 6
MD intervention determines a reduction of faecal branched-chain fatty acid (BCFA) concentrations and higher levels of Faecalibacterium prausnitzii and Lachnospiraceae taxa. Parallel coordinate plot showing variations of (A) valerate, (B) isovalerate, (C) isobutyrate and (D) 2-methylbutyrate faecal concentrations within MedD population. The red triangles indicate mean values, the lines connecting dots are used to indicate the same sample at each time point. The significance was tested by applying the post hoc Friedman-Nemenyi test for pairwise test of multiple comparisons within each group. In the box plots, the relative abundances of (E) F. prausnitzii 3 and (F) Lachnospiraceae family are compared considering subjects falling in the highest quartile (n=16, violet) and in the lowest quartile (n=16, purple) of faecal butyrate increase after 4 weeks of treatment. Statistical differences between groups were determined using Wilcoxon rank-sum tests. Baseline, 0 weeks; 4w, 4 weeks; 8w, 8 weeks of intervention. H, highest quartile of increase; L, lowest quartile of increase.
Figure 2
Figure 2
Mediterranean diet changes the intestinal and systemic metabolome. Partial least squares discriminant analysis plots based on molecular features detected in (A) faeces and (B) urine. Subjects belonging to different categories were coloured according to diet and time points: MedD subjects at baseline (light green), after 4 weeks (green) and 8 weeks of intervention (dark green). ConD subjects at baseline (light orange), after 4 weeks (orange) and 8 weeks (dark orange) of intervention. The loading plots display vectors that contributed the most to variability of individual dataset; variables explaining the variance between the groups in (C) faecal and (D) urine metabolome are reported as bar plots.
Figure 3
Figure 3
Microbial diversity richness anticorrelates with inflammation. (A) Spearman’s correlation between variation of gut microbial gene richness and individual inflammatory status (serum hs-CRP) variation at the end of trial; n observation=62. (B) Violin plot showing differences in serum hs-CRP variation between subjects increasing (n=25, yellow) compared with subjects decreasing (n=37, light blue) gene richness at the end of trial. Statistical differences between groups were determined using unpaired Wilcoxon rank-sum tests. hs-CRP, high sensitivity C reactive protein.
Figure 4
Figure 4
Mediterranean diet (MD) affects gut microbiome composition. (A) Total delta MD index changes over the 4w-baseline period. Left: histograms of delta (4w-baseline) MD index (n=62). Right: linear regressions of microbiome similarity compared with delta 4w-baseline MD index. Microbiome similarity was estimated by Spearman correlations between microbial composition at 4 weeks and baseline within each individual. (B) Total MD index fractional changes (FCs) (4w-baseline)/baseline, used as proxy to measure the effort of adherence (n=62). Left: distribution of individuals relative to MD index FC. Right: linear regressions of microbiome similarity and FC. Spearman correlations (rho and p values) are reported, excluding outliers, for ConD (n=31) and MedD (n=26) groups, respectively. 4w, 4 weeks.
Figure 5
Figure 5
Faecal BA concentrations over the nutritional intervention. Parallel coordinate plot showing variations of faecal (A) total, (B) primary and (C) secondary BA concentrations within the MedD group during the intervention. The red triangles indicate mean values, the lines connecting dots are used to indicate the same sample at each time point. The significance was tested by applying the post hoc Friedman-Nemenyi test for pairwise test of multiple comparisons within each group. (D) In the box plot the relative abundances of Bilophila wadsworthia are compared considering subjects falling in the highest quartile (n=16, green) and in the lowest quartile of reduction (n=16, blue) of secondary to primary BAs ratio after 4 weeks of treatment. Baseline, 0 weeks; 4w, 4 weeks; 8w, 8 weeks of nutritional intervention. H, highest quartile of reduction; L, lowest quartile of reduction; BAs, bile acids.
Figure 6
Figure 6
MD intervention determines a reduction of faecal branched-chain fatty acid (BCFA) concentrations and higher levels of Faecalibacterium prausnitzii and Lachnospiraceae taxa. Parallel coordinate plot showing variations of (A) valerate, (B) isovalerate, (C) isobutyrate and (D) 2-methylbutyrate faecal concentrations within MedD population. The red triangles indicate mean values, the lines connecting dots are used to indicate the same sample at each time point. The significance was tested by applying the post hoc Friedman-Nemenyi test for pairwise test of multiple comparisons within each group. In the box plots, the relative abundances of (E) F. prausnitzii 3 and (F) Lachnospiraceae family are compared considering subjects falling in the highest quartile (n=16, violet) and in the lowest quartile (n=16, purple) of faecal butyrate increase after 4 weeks of treatment. Statistical differences between groups were determined using Wilcoxon rank-sum tests. Baseline, 0 weeks; 4w, 4 weeks; 8w, 8 weeks of intervention. H, highest quartile of increase; L, lowest quartile of increase.

References

    1. Kelly JT, Palmer SC, Wai SN, et al. . Healthy dietary patterns and risk of mortality and ESRD in CKD: a meta-analysis of cohort studies. Clin J Am Soc Nephrol 2017;12:272–9. 10.2215/CJN.06190616
    1. Chiavaroli L, Viguiliouk E, Nishi SK, et al. . Dash dietary pattern and cardiometabolic outcomes: an umbrella review of systematic reviews and meta-analyses. Nutrients 2019;11:338 10.3390/nu11020338
    1. Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res 2019;124:779–98. 10.1161/CIRCRESAHA.118.313348
    1. Bendall CL, Mayr HL, Opie RS, et al. . Central obesity and the Mediterranean diet: a systematic review of intervention trials. Crit Rev Food Sci Nutr 2018;58:3070–84. 10.1080/10408398.2017.1351917
    1. Eleftheriou D, Benetou V, Trichopoulou A, et al. . Mediterranean diet and its components in relation to all-cause mortality: meta-analysis. Br J Nutr 2018;120:1081–97. 10.1017/S0007114518002593
    1. Cani PD. Human gut microbiome: hopes, threats and promises. Gut 2018;67:1716–25. 10.1136/gutjnl-2018-316723
    1. David LA, Maurice CF, Carmody RN, et al. . Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505:559–63. 10.1038/nature12820
    1. Roager HM, Dragsted LO. Diet‐derived microbial metabolites in health and disease. Nutr Bull 2019;44:216–27. 10.1111/nbu.12396
    1. Johnson AJ, Vangay P, Al-Ghalith GA, et al. . Daily sampling reveals personalized Diet-Microbiome associations in humans. Cell Host Microbe 2019;25:789–802. 10.1016/j.chom.2019.05.005
    1. Bashiardes S, Godneva A, Elinav E, et al. . Towards utilization of the human genome and microbiome for personalized nutrition. Curr Opin Biotechnol 2018;51:57–63. 10.1016/j.copbio.2017.11.013
    1. Grosso G, Micek A, Godos J, et al. . Health risk factors associated with meat, fruit and vegetable consumption in cohort studies: a comprehensive meta-analysis. PLoS One 2017;12:e0183787 10.1371/journal.pone.0183787
    1. De Filippis F, Vitaglione P, Cuomo R, et al. . Dietary interventions to modulate the gut Microbiome-How far away are we from precision medicine. Inflamm Bowel Dis 2018;24:2142–54. 10.1093/ibd/izy080
    1. Bailey MA, Holscher HD. Microbiome-mediated effects of the Mediterranean diet on inflammation. Adv Nutr 2018;9:193–206. 10.1093/advances/nmy013
    1. Harris RP, Helfand M, Woolf SH, et al. . Current methods of the US preventive services Task force: a review of the process. Am J Prev Med 2001;20:21–35. 10.1016/s0749-3797(01)00261-6
    1. Blumberg J, Heaney RP, Huncharek M, et al. . Evidence-Based criteria in the nutritional context. Nutr Rev 2010;68:478–84. 10.1111/j.1753-4887.2010.00307.x
    1. Myles IA. Fast food fever: reviewing the impacts of the Western diet on immunity. Nutr J 2014;13:61 10.1186/1475-2891-13-61
    1. Minihane AM, Vinoy S, Russell WR, et al. . Low-Grade inflammation, diet composition and health: current research evidence and its translation. Br J Nutr 2015;114:999–1012. 10.1017/S0007114515002093
    1. Mozaffarian D, Hao T, Rimm EB, et al. . Changes in diet and lifestyle and long-term weight gain in women and men. N Engl J Med 2011;364:2392–404. 10.1056/NEJMoa1014296
    1. Agnoli C, Krogh V, Grioni S, et al. . A priori-defined dietary patterns are associated with reduced risk of stroke in a large Italian cohort. J Nutr 2011;141:1552–8. 10.3945/jn.111.140061
    1. Andersen M-BS, Kristensen M, Manach C, et al. . Discovery and validation of urinary exposure markers for different plant foods by untargeted metabolomics. Anal Bioanal Chem 2014;406:1829–44. 10.1007/s00216-013-7498-5
    1. Barri T, Dragsted LO. UPLC-ESI-QTOF/MS and multivariate data analysis for blood plasma and serum metabolomics: effect of experimental artefacts and anticoagulant. Anal Chim Acta 2013;768:118–28. 10.1016/j.aca.2013.01.015
    1. Christiansen CB, Gabe MBN, Svendsen B, et al. . The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. Am J Physiol Gastrointest Liver Physiol 2018;315:G53–65. 10.1152/ajpgi.00346.2017
    1. Hjerpsted JB, Dragsted LO, Tholstrup T. Cheese intake lowers plasma cholesterol concentrations without increasing bile acid excretion. J Nutr Intermed Metab 2016;3:12–17. 10.1016/j.jnim.2015.11.002
    1. Plaza Oñate F, Le Chatelier E, Almeida M, et al. . MSPminer: abundance-based reconstitution of microbial pan-genomes from shotgun metagenomic data. Bioinformatics 2019;35:1544–52. 10.1093/bioinformatics/bty830
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 2016;32:2847–9. 10.1093/bioinformatics/btw313
    1. Pasolli E, Truong DT, Malik F, et al. . Machine learning meta-analysis of large metagenomic datasets: tools and biological insights. PLoS Comput Biol 2016;12:e1004977 10.1371/journal.pcbi.1004977
    1. Drescher LS, Thiele S, Mensink GBM. A new index to measure healthy food diversity better reflects a healthy diet than traditional measures. J Nutr 2007;137:647–51. 10.1093/jn/137.3.647
    1. Roager HM, Vogt JK, Kristensen M, et al. . Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial. Gut 2019;68:83–93. 10.1136/gutjnl-2017-314786
    1. Keller BO, Wu BTF, Li SSJ, et al. . Hypaphorine is present in human milk in association with consumption of legumes. J Agric Food Chem 2013;61:7654–60. 10.1021/jf401758f
    1. Cuparencu CS, Andersen M-BS, Gürdeniz G, et al. . Identification of urinary biomarkers after consumption of sea buckthorn and strawberry, by untargeted LC–MS metabolomics: a meal study in adult men. Metabolomics 2016;12:31 10.1007/s11306-015-0934-0
    1. Tulipani S, Urpi-Sarda M, García-Villalba R, et al. . Urolithins are the main urinary microbial-derived phenolic metabolites discriminating a moderate consumption of nuts in free-living subjects with diagnosed metabolic syndrome. J Agric Food Chem 2012;60:8930–40. 10.1021/jf301509w
    1. Dragsted LO. Biomarkers of meat intake and the application of nutrigenomics. Meat Sci 2010;84:301–7. 10.1016/j.meatsci.2009.08.028
    1. Khakimov B, Poulsen SK, Savorani F, et al. . New Nordic diet versus average Danish diet: a randomized controlled trial revealed healthy long-term effects of the new Nordic diet by GC-MS blood plasma metabolomics. J Proteome Res 2016;15:1939–54. 10.1021/acs.jproteome.6b00109
    1. Singh A, Shannon CP, Gautier B, et al. . DIABLO: an integrative approach for identifying key molecular drivers from multi-omics assays. Bioinformatics 2019;35:3055–62. 10.1093/bioinformatics/bty1054
    1. Lewington S, Whitlock G, Clarke R, et al. . Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths. Lancet 2007;370:1829–39. 10.1016/S0140-6736(07)61778-4
    1. Guasch-Ferré M, Satija A, Blondin SA, et al. . Meta-Analysis of randomized controlled trials of red meat consumption in comparison with various comparison diets on cardiovascular risk factors. Circulation 2019;139:1828–45. 10.1161/CIRCULATIONAHA.118.035225
    1. García-Conesa M-T, Chambers K, Combet E, et al. . Meta-Analysis of the effects of foods and derived products containing ellagitannins and anthocyanins on cardiometabolic biomarkers: analysis of factors influencing variability of the individual responses. Int J Mol Sci 2018;19:694 10.3390/ijms19030694
    1. Wolff E, Vergnes M-F, Portugal H, et al. . Cholesterol-absorber status modifies the LDL cholesterol-lowering effect of a Mediterranean-type diet in adults with moderate cardiovascular risk factors. J Nutr 2011;141:1791–8. 10.3945/jn.111.141333
    1. Bouchard-Mercier A, Rudkowska I, Lemieux S, et al. . The metabolic signature associated with the Western dietary pattern: a cross-sectional study. Nutr J 2013;12:158 10.1186/1475-2891-12-158
    1. Schmidt JA, Rinaldi S, Ferrari P, et al. . Metabolic profiles of male meat eaters, fish eaters, vegetarians, and vegans from the EPIC-Oxford cohort. Am J Clin Nutr 2015;102:1518–26. 10.3945/ajcn.115.111989
    1. Ross AB, Pere-Trépat E, Montoliu I, et al. . A whole-grain-rich diet reduces urinary excretion of markers of protein catabolism and gut microbiota metabolism in healthy men after one week. J Nutr 2013;143:766–73. 10.3945/jn.112.172197
    1. Guasch-Ferré M, Zheng Y, Ruiz-Canela M, et al. . Plasma acylcarnitines and risk of cardiovascular disease: effect of Mediterranean diet interventions. Am J Clin Nutr 2016;103:1408–16. 10.3945/ajcn.116.130492
    1. Korver RA, Koevoets IT, Testerink C. Out of shape during stress: a key role for auxin. Trends Plant Sci 2018;23:783–93. 10.1016/j.tplants.2018.05.011
    1. Chávez-Talavera O, Tailleux A, Lefebvre P, et al. . Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 2017;152:1679–94. 10.1053/j.gastro.2017.01.055
    1. Draper CF, Vassallo I, Di Cara A, et al. . A 48-hour vegan diet challenge in healthy women and men induces a branch-chain amino acid related, health associated, metabolic signature. Mol Nutr Food Res 2018;62:1700703 10.1002/mnfr.201700703
    1. De Filippis F, Pellegrini N, Vannini L, et al. . High-Level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016;65:1812–21. 10.1136/gutjnl-2015-309957
    1. de la Cuesta-Zuluaga J, Mueller N, Álvarez-Quintero R, et al. . Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk factors. Nutrients 2019;11:51 10.3390/nu11010051
    1. Mitsou EK, Kakali A, Antonopoulou S, et al. . Adherence to the Mediterranean diet is associated with the gut microbiota pattern and gastrointestinal characteristics in an adult population. Br J Nutr 2017;117:1645–55. 10.1017/S0007114517001593
    1. Cerdá B, Periago P, Espín JC, et al. . Identification of urolithin a as a metabolite produced by human colon microflora from ellagic acid and related compounds. J Agric Food Chem 2005;53:5571–6. 10.1021/jf050384i
    1. Garcia-Aloy M, Hulshof PJM, Estruel-Amades S, et al. . Biomarkers of food intake for nuts and vegetable oils: an extensive literature search. Genes Nutr 2019;14:7 10.1186/s12263-019-0628-8
    1. Singh R, Chandrashekharappa S, Bodduluri SR, et al. . Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat Commun 2019;10:89 10.1038/s41467-018-07859-7
    1. Selma MV, González-Sarrías A, Salas-Salvadó J, et al. . The gut microbiota metabolism of pomegranate or walnut ellagitannins yields two urolithin-metabotypes that correlate with cardiometabolic risk biomarkers: comparison between normoweight, overweight-obesity and metabolic syndrome. Clin Nutr 2018;37:897–905. 10.1016/j.clnu.2017.03.012
    1. Stanisławska IJ, Granica S, Piwowarski JP, et al. . The activity of urolithin A and M4 Valerolactone, colonic microbiota metabolites of polyphenols, in a prostate cancer in vitro model. Planta Med 2019;85:118–25. 10.1055/a-0755-7715
    1. Zhang W, Chen J-H, Aguilera-Barrantes I, et al. . Urolithin a suppresses the proliferation of endometrial cancer cells by mediating estrogen receptor-α-dependent gene expression. Mol Nutr Food Res 2016;60:2387–95. 10.1002/mnfr.201600048
    1. Teixeira LL, Costa GR, Dörr FA, et al. . Potential antiproliferative activity of polyphenol metabolites against human breast cancer cells and their urine excretion pattern in healthy subjects following acute intake of a polyphenol-rich juice of grumixama (Eugenia brasiliensis Lam.). Food Funct 2017;8:2266–74. 10.1039/C7FO00076F
    1. Haro C, Montes-Borrego M, Rangel-Zúñiga OA, et al. . Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population. J Clin Endocrinol Metab 2016;101:233–42. 10.1210/jc.2015-3351
    1. Haro C, Garcia-Carpintero S, Alcala-Diaz JF, et al. . The gut microbial community in metabolic syndrome patients is modified by diet. J Nutr Biochem 2016;27:27–31. 10.1016/j.jnutbio.2015.08.011
    1. Vanegas SM, Meydani M, Barnett JB, et al. . 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;105:635–50. 10.3945/ajcn.116.146928
    1. Vitaglione P, Mennella I, Ferracane R, 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;101:251–61. 10.3945/ajcn.114.088120
    1. Henke MT, Kenny DJ, Cassilly CD, et al. . Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn's disease, produces an inflammatory polysaccharide. Proc Natl Acad Sci U S A 2019;116:12672–7. 10.1073/pnas.1904099116
    1. Le Chatelier E, Nielsen T, Qin J, et al. . Richness of human gut microbiome correlates with metabolic markers. Nature 2013;500:541–6. 10.1038/nature12506
    1. Brahe LK, Le Chatelier E, Prifti E, et al. . Specific gut microbiota features and metabolic markers in postmenopausal women with obesity. Nutr Diabetes 2015;5:e159 10.1038/nutd.2015.9
    1. Devkota S, Wang Y, Musch MW, et al. . Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in IL10-/- mice. Nature 2012;487:104–8. 10.1038/nature11225
    1. Natividad JM, Lamas B, Pham HP, et al. . Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat Commun 2018;9:2802 10.1038/s41467-018-05249-7
    1. Sokol H, Pigneur B, Watterlot L, et al. . Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008;105:16731–6. 10.1073/pnas.0804812105
    1. Vital M, Karch A, Pieper DH. Colonic butyrate-producing communities in humans: an overview using omics data. mSystems 2017;2:e00130–17. 10.1128/mSystems.00130-17
    1. Pedersen HK, Gudmundsdottir V, Nielsen HB, et al. . Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016;535:376–81. 10.1038/nature18646
    1. De Filippis F, Pasolli E, Tett A, et al. . Distinct genetic and functional traits of human intestinal Prevotella copri strains are associated with different habitual diets. Cell Host Microbe 2019;25:444–53. 10.1016/j.chom.2019.01.004

Source: PubMed

3
購読する