Protein supplementation during an energy-restricted diet induces visceral fat loss and gut microbiota amino acid metabolism activation: a randomized trial

Pierre Bel Lassen, Eugeni Belda, Edi Prifti, Maria Carlota Dao, Florian Specque, Corneliu Henegar, Laure Rinaldi, Xuedan Wang, Sean P Kennedy, Jean-Daniel Zucker, Wim Calame, Benoît Lamarche, Sandrine P Claus, Karine Clément, Pierre Bel Lassen, Eugeni Belda, Edi Prifti, Maria Carlota Dao, Florian Specque, Corneliu Henegar, Laure Rinaldi, Xuedan Wang, Sean P Kennedy, Jean-Daniel Zucker, Wim Calame, Benoît Lamarche, Sandrine P Claus, Karine Clément

Abstract

Interactions between diet and gut microbiota are critical regulators of energy metabolism. The effects of fibre intake have been deeply studied but little is known about the impact of proteins. Here, we investigated the effects of high protein supplementation (Investigational Product, IP) in a double blind, randomised placebo-controled intervention study (NCT01755104) where 107 participants received the IP or an isocaloric normoproteic comparator (CP) alongside a mild caloric restriction. Gut microbiota profiles were explored in a patient subset (n = 53) using shotgun metagenomic sequencing. Visceral fat decreased in both groups (IP group: - 20.8 ± 23.2 cm2; CP group: - 14.5 ± 24.3 cm2) with a greater reduction (p < 0.05) with the IP supplementation in the Per Protocol population. Microbial diversity increased in individuals with a baseline low gene count (p < 0.05). The decrease in weight, fat mass and visceral fat mass significantly correlated with the increase in microbial diversity (p < 0.05). Protein supplementation had little effects on bacteria composition but major differences were seen at functional level. Protein supplementation stimulated bacterial amino acid metabolism (90% amino-acid synthesis functions enriched with IP versus 13% in CP group (p < 0.01)). Protein supplementation alongside a mild energy restriction induces visceral fat mass loss and an activation of gut microbiota amino-acid metabolism.Clinical trial registration: NCT01755104 (24/12/2012). https://ichgcp.net/clinical-trials-registry/NCT01755104?term=NCT01755104&draw=2&rank=1 .

Conflict of interest statement

SPC works as Chief Scientific Officer at YSOPIA Bioscience (Bordeaux, France). LR works as project manager at YSOPIA Bioscience (Bordeaux, France). KC is currently a member of the Scientific Advisory board of YSOPIA Bioscience but this activity is not related to the topic of this publication. All the other authors declare no conflict of interest.

© 2021. The Author(s).

Figures

Figure 1
Figure 1
Investigational Product improved fat mass, maintained lean mass and reduced inflammation at week 12 post intervention. Changes in visceral fat area as established via CT scan in the (A) FAS population, (B) FAS without outliers and (C) PP population. Changes in fat mass (D) and lean mass (E) as established via DEXA scan and in TNFα (F) in FAS without outliers. Mean (SEM) for at least n = 51. *p < 0.05 (GEE model) Key: Circle is Control Product; Square is Investigational Product.
Figure 2
Figure 2
Evolution of metagenomic richness 12 weeks after dietary intervention. (A) Evolution of gene count (number of genes) and metagenomic richness (number of MGS) depending on baseline gene richness status The fixed effect of time (the intervention) was analysed in a mixed linear model with patients as random effects. *p < 0.05, **p < 0.001. (B) Evolution of richness associations with the clinical evolution. Heatmap of standardized beta coefficient from linear regression. Model is adjusted for baseline values + baseline BMI and sex. For cholesterol and triglycerides, the model is also adjusted for baseline statin intake. (C) Evolution of weight, fat mass, visceral fat mass and gene count depending on richness response. Change from baseline is (T12–T0/)T0 * 100. Gained species are individuals who increased their MGS richness. Lost species are individuals who decreased their metagenomic (MGS) richness. P values of the effect of MGS richness change in a linear regression model adjusted for baseline value, sex and BMI. Points are mean, bars are SEM. Figure conceived using R version 3.3.2, R Core Team (2019), https://www.R-project.org/.
Figure 3
Figure 3
Effects of protein supplementation on gut microbiome composition (IP vs. CP). (A) Evolution (relative change at T12) of gene count and MGS richness (number of present species) in investigational product group (IP, red colours) and Comparator group (CP, blue colours) depending on baseline metagenomic richness. LGC: low gene count at baseline (light colours). HGC: high gene count at baseline. p value of the effect intervention interaction with time in LGC patients (a) and HGC patients (b); (B) Alluvial plot showing the evolution of enterotype between T0 and T12 in CP group (left panel) and IP group (right panel); (C) Untargeted analysis of the effects of IP on metagenomic species (MGS) abundance changes with the intervention. MGS shown are the ones with a significant interaction of time with intervention in a mixed linear model with patients as random effects adjusted for baseline age, sex and BMI (p < 0.05 without adjustment for multiple comparisons). Bars represent the cliff delta effect of time (T12 vs T0) on MGS abundance in each group. *p < 0.05; **p < 0.01; ***p < 0.001 No significant differences resist to adjustment for multiple comparisons. IP: investigational product (high protein); CP: comparator product. Figure conceived using R version 3.3.2, R Core Team (2019), https://www.R-project.org/.
Figure 4
Figure 4
Effects of protein supplementation on gut microbiota function. (A) Effects of IP on functional modules from KEGG and GMM databases. Shown modules are those which changed differently (FDR < 0.1) between the two groups (IP vs. CP) adjusted for baseline BMI, age and sex. Bars represent log(fold change) at T12 of modules abundances in each group (IP vs. CP). Modules in bold: amino-acid metabolism modules. *p < 0.05; **p < 0.01; ***p < 0.001. (B) Heatmap of spearman correlations between functional modules fold changes and bio-clinical relative changes after the intervention (T12-T0/T0); *p < 0.05; **q < 0.05 (adjusted for multiple comparison, FDR method). (C) Evolution of the amino acid metabolism (degradation and synthesis) functional modules (KEGG and GMM) with the intervention. Increase is defined by a mean fold change > 0 and decrease by a mean fold change < 0 for each module. The observed proportion of increased module was compared to a theoretical value of 0.5 with a binomial test. *p < 0.05; **p < 0.01; ***p < 0.001 (adjusted for multiple comparisons, FDR method). (D) Effects of IP on amino acid metabolism functional groups of metagenomic species. Shown functional groups are those which changed differently (p < 0.05) between the two groups (IP vs. CP) adjusted for baseline BMI, age and sex. Bars represent log(fold change) at T12 of modules abundances in each group (IP vs. CP). *p < 0.05; **p < 0.01; ***p < 0.001. IP: investigational product (high protein); CP: comparator product. Figure conceived using R version 3.3.2, R Core Team (2019), https://www.R-project.org/.

References

    1. De Lorenzo A, et al. Adiposity rather than BMI determines metabolic risk. Int. J. Cardiol. 2013;166:111–117. doi: 10.1016/j.ijcard.2011.10.006.
    1. Calder PC, et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011;106(Suppl 3):S5–78. doi: 10.1017/S0007114511005460.
    1. Mottillo S, et al. The metabolic syndrome and cardiovascular risk: A systematic review and meta-analysis. J. Am. Coll. Cardiol. 2010;56:1113–1132. doi: 10.1016/j.jacc.2010.05.034.
    1. Lorenzo C, Okoloise M, Williams K, Stern MP, Haffner SM. The metabolic syndrome as predictor of type 2 diabetes: The San Antonio Heart Study. Diabetes Care. 2003;26:3153–3159. doi: 10.2337/diacare.26.11.3153.
    1. Merlotti C, Ceriani V, Morabito A, Pontiroli AE. Subcutaneous fat loss is greater than visceral fat loss with diet and exercise, weight-loss promoting drugs and bariatric surgery: A critical review and meta-analysis. Int. J. Obes. 2017;2005(41):672–682. doi: 10.1038/ijo.2017.31.
    1. Le Roy CI, et al. Dissecting the role of the gut microbiota and diet on visceral fat mass accumulation. Sci. Rep. 2019;9:9758. doi: 10.1038/s41598-019-46193-w.
    1. Nie X, et al. A metagenome-wide association study of gut microbiome and visceral fat accumulation. Comput. Struct. Biotechnol. J. 2020;18:2596–2609. doi: 10.1016/j.csbj.2020.09.026.
    1. Campbell WW, Kim JE, Amankwaah AF, Gordon SL, Weinheimer-Haus EM. Higher total protein intake and change in total protein intake affect body composition but not metabolic syndrome indexes in middle-aged overweight and obese adults who perform resistance and aerobic exercise for 36 weeks. J. Nutr. 2015;145:2076–2083. doi: 10.3945/jn.115.213595.
    1. Huang G, et al. Effect of protein intake on visceral abdominal fat and metabolic biomarkers in older men with functional limitations: Results from a randomized clinical trial. J. Gerontol. A. Biol. Sci. Med. Sci. 2021 doi: 10.1093/gerona/glab007.
    1. Takahira M, et al. Randomized, double-blind, controlled, comparative trial of formula food containing soy protein vs. milk protein in visceral fat obesity. FLAVO study. Circ. J. Off. J. Jpn. Circ. Soc. 2011;75:2235–2243.
    1. Blachier F, et al. High-protein diets for weight management: Interactions with the intestinal microbiota and consequences for gut health. A position paper by the my new gut study group. Clin. Nutr. 2018 doi: 10.1016/j.clnu.2018.09.016.
    1. Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64. doi: 10.1038/nature18846.
    1. Wu GD, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–108. doi: 10.1126/science.1208344.
    1. Zhernakova A, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–569. doi: 10.1126/science.aad3369.
    1. Liu R, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat. Med. 2017;23:859–868. doi: 10.1038/nm.4358.
    1. Koh A, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175:947–961.e17. doi: 10.1016/j.cell.2018.09.055.
    1. Karlsson FH, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498:99–103. doi: 10.1038/nature12198.
    1. Aron-Wisnewsky J, Clément K. The gut microbiome, diet and links to cardiometabolic and chronic disorders. Nat. Rev. Nephrol. 2016;12:169–181. doi: 10.1038/nrneph.2015.191.
    1. Cotillard A, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500:585–588. doi: 10.1038/nature12480.
    1. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi: 10.1038/nature12820.
    1. Walker AW, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5:220–230. doi: 10.1038/ismej.2010.118.
    1. Salonen A, et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014;8:2218–2230. doi: 10.1038/ismej.2014.63.
    1. Dong TS, et al. A high protein calorie restriction diet alters the gut microbiome in obesity. Nutrients. 2020;12:3221. doi: 10.3390/nu12103221.
    1. Alberti KGM, Zimmet P, Shaw J. The metabolic syndrome—A new worldwide definition. Lancet. 2005;366:1059–1062. doi: 10.1016/S0140-6736(05)67402-8.
    1. Labonté M-È, Cyr A, Baril-Gravel L, Royer M-M, Lamarche B. Validity and reproducibility of a web-based, self-administered food frequency questionnaire. Eur. J. Clin. Nutr. 2012;66:166–173. doi: 10.1038/ejcn.2011.163.
    1. Nielsen HB, et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat. Biotechnol. 2014;32:822–828. doi: 10.1038/nbt.2939.
    1. Holmes I, Harris K, Quince C. Dirichlet multinomial mixtures: Generative models for microbial metagenomics. PLoS ONE. 2012;7:e30126. doi: 10.1371/journal.pone.0030126.
    1. Abubucker S, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 2012;8:e1002358. doi: 10.1371/journal.pcbi.1002358.
    1. Vieira-Silva S, et al. Species–function relationships shape ecological properties of the human gut microbiome. Nat. Microbiol. 2016;1:16088. doi: 10.1038/nmicrobiol.2016.88.
    1. Darzi Y, Falony G, Vieira-Silva S, Raes J. Towards biome-specific analysis of meta-omics data. ISME J. 2016;10:1025–1028. doi: 10.1038/ismej.2015.188.
    1. Oksanen, J. et al. vegan: Community Ecology Package (2020).
    1. Ajala O, English P, Pinkney J. Systematic review and meta-analysis of different dietary approaches to the management of type 2 diabetes. Am. J. Clin. Nutr. 2013;97:505–516. doi: 10.3945/ajcn.112.042457.
    1. Leidy HJ, Carnell NS, Mattes RD, Campbell WW. Higher Protein Intake Preserves Lean Mass and Satiety with Weight Loss in Pre-obese and Obese Women. Obesity. 2007;15:421–429. doi: 10.1038/oby.2007.531.
    1. Piatti PM, et al. Hypocaloric high-protein diet improves glucose oxidation and spares lean body mass: Comparison to hypocaloric high-carbohydrate diet. Metabolism. 1994;43:1481–1487. doi: 10.1016/0026-0495(94)90005-1.
    1. Noakes M, Keogh JB, Foster PR, Clifton PM. Effect of an energy-restricted, high-protein, low-fat diet relative to a conventional high-carbohydrate, low-fat diet on weight loss, body composition, nutritional status, and markers of cardiovascular health in obese women. Am. J. Clin. Nutr. 2005;81:1298–1306. doi: 10.1093/ajcn/81.6.1298.
    1. Farnsworth E, et al. Effect of a high-protein, energy-restricted diet on body composition, glycemic control, and lipid concentrations in overweight and obese hyperinsulinemic men and women. Am. J. Clin. Nutr. 2003;78:31–39. doi: 10.1093/ajcn/78.1.31.
    1. Le Chatelier E, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546. doi: 10.1038/nature12506.
    1. Aron-Wisnewsky J, et al. Major microbiota dysbiosis in severe obesity: Fate after bariatric surgery. Gut. 2018 doi: 10.1136/gutjnl-2018-316103.
    1. Vandeputte D, et al. Quantitative microbiome profiling links gut community variation to microbial load. Nature. 2017;551:507–511. doi: 10.1038/nature24460.
    1. Beaumont M, et al. Quantity and source of dietary protein influence metabolite production by gut microbiota and rectal mucosa gene expression: A randomized, parallel, double-blind trial in overweight humans. Am. J. Clin. Nutr. 2017;106:1005–1019. doi: 10.3945/ajcn.117.158816.
    1. Natividad JM, et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 2018;9:2802. doi: 10.1038/s41467-018-05249-7.
    1. Windey K, et al. Modulation of protein fermentation does not affect fecal water toxicity: A randomized cross-over study in healthy subjects. PLoS ONE. 2012;7:e52387. doi: 10.1371/journal.pone.0052387.
    1. Muegge BD, et al. diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science. 2011;332:970–974. doi: 10.1126/science.1198719.
    1. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222. doi: 10.1038/nature11053.
    1. Massey LK, Sokatch JR, Conrad RS. Branched-chain amino acid catabolism in bacteria. Bacteriol. Rev. 1976;40:42–54. doi: 10.1128/br.40.1.42-54.1976.
    1. Rudman D, Meister A. Transamination in Escherichia coli. J. Biol. Chem. 1953;200:591–604. doi: 10.1016/S0021-9258(18)71404-X.
    1. Fouillet H, et al. Urea-nitrogen production and salvage are modulated by protein intake in fed humans: Results of an oral stable-isotope-tracer protocol and compartmental modeling. Am. J. Clin. Nutr. 2008;87:1702–1714. doi: 10.1093/ajcn/87.6.1702.
    1. Beaumont M, et al. Structure of protein emulsion in food impacts intestinal microbiota, caecal luminal content composition and distal intestine characteristics in rats. Mol. Nutr. Food Res. 2017;61:1700078. doi: 10.1002/mnfr.201700078.

Source: PubMed

3
Abonnieren