The Metabolic Response to a Low Amino Acid Diet is Independent of Diet-Induced Shifts in the Composition of the Gut Microbiome

Heidi H Pak, Nicole E Cummings, Cara L Green, Jacqueline A Brinkman, Deyang Yu, Jay L Tomasiewicz, Shany E Yang, Colin Boyle, Elizabeth N Konon, Irene M Ong, Dudley W Lamming, Heidi H Pak, Nicole E Cummings, Cara L Green, Jacqueline A Brinkman, Deyang Yu, Jay L Tomasiewicz, Shany E Yang, Colin Boyle, Elizabeth N Konon, Irene M Ong, Dudley W Lamming

Abstract

Obesity and type 2 diabetes are increasing in prevalence around the world, and there is a clear need for new and effective strategies to promote metabolic health. A low protein (LP) diet improves metabolic health in both rodents and humans, but the mechanisms that underlie this effect remain unknown. The gut microbiome has recently emerged as a potent regulator of host metabolism and the response to diet. Here, we demonstrate that a LP diet significantly alters the taxonomic composition of the gut microbiome at the phylum level, altering the relative abundance of Actinobacteria, Bacteroidetes, and Firmicutes. Transcriptional profiling suggested that any impact of the microbiome on liver metabolism was likely independent of the microbiome-farnesoid X receptor (FXR) axis. We therefore tested the ability of a LP diet to improve metabolic health following antibiotic ablation of the gut microbiota. We found that a LP diet promotes leanness, increases energy expenditure, and improves glycemic control equally well in mice treated with antibiotics as in untreated control animals. Our results demonstrate that the beneficial effects of a LP diet on glucose homeostasis, energy balance, and body composition are unlikely to be mediated by diet-induced changes in the taxonomic composition of the gut microbiome.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
A low protein diet promotes metabolic health and alters the taxonomic composition of the cecal microbiome. (A) Glucose tolerance test on male C57BL/6J mice fed a Control (22% of calories from amino acids) or Low AA (7% of calories from amino acids) diet for 4 months (n = 8–10/group; *p < 0.05, t-test). (B) Weight and body composition were measured immediately prior to diet start and after 10 weeks on the indicated diets (n = 8–10/group; *p < 0.05, = t-test). (C) Bar plot of average relative abundance at the phylum taxonomic level. Top 6 phyla are shown. (D) Principle component analysis of demonstrating the effect of diet on taxonomic composition. (E,F) Bacterial phyla differentially represented in cecal contents from mice fed the specified diets for 4 months (n = 7–12/group; Sidak’s test following ANOVA, *p < 0.05). Error bars represent SEM.
Figure 2
Figure 2
A low protein diet alters the hepatic transcriptome and shows distinct changes in biological pathways. (AC) RNA-Seq was performed on the livers for mice fed a Control diet or a Low AA for four months. (A) A heatmap indicating the relative expression of genes involved in the most significantly enriched biological KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways based on genes differentially expressed in the livers of Control and Low AA fed mice (q < 0.05, FDR). Genes in more than one significantly enriched KEGG pathway are listed only once, and assigned to the most significantly affected pathway. (B) Pathway enrichment analysis was performed using g:Profiler (g:GOSt), and the p-values of KEGG pathways significantly up- and downregulated by Low AA diet feeding were determined. Colors are matched to that of pathways in (A). (C) Heatmap representing the relative expression of liver genes known to be altered by FXR-FGF15 bile acid signaling.
Figure 3
Figure 3
A low protein diet alters body composition similarly in vehicle and antibiotic-treated mice. (A) Schematic representation of the experimental plan; mice were pretreated with antibiotics or vehicle for three weeks, and then randomized to either a Control or Low AA diet. (B) Fecal DNA content was determined following 3 weeks of antibiotic treatment (n = 8/group; *p < 0.05, t-test). (C) Weight of the mice in each group was tracked following randomization to each diet. (DF) Weight and body composition were determined immediately prior to diet start and after 6 weeks on the indicated diets, and the change in (D) weight, (E) fat mass, and (F) lean mass was determined (n = 12/group; statistics for the overall effects of diet, antibiotic (ABX) treatment, and the interaction represent the p-value from a two-way ANOVA; *p < 0.05 from a Sidak’s post-test examining the effect of parameters identified as significant in the two-way ANOVA). Error bars represent SEM.
Figure 4
Figure 4
A low protein diet improves glucose homeostasis similarly in vehicle and antibiotic-treated mice. (A) Glucose and (B) alanine tolerance tests were conducted in mice fed the indicated diets for 8 weeks and 4 weeks, respectively (n = 12/group; statistics for the overall effects of diet, antibiotic (ABX) treatment, and the interaction represent the p-value from a two-way ANOVA; *p < 0.05 from a Sidak’s post-test examining the effect of parameters identified as significant in the two-way ANOVA). Error bars represent SEM.
Figure 5
Figure 5
A low protein diet increases food consumption and energy expenditure similarly in vehicle and antibiotic-treated mice. (AF) Metabolic chambers were used to assess (A,B) food consumption, (C) spontaneous activity, (D) respiratory exchange ratio (RER), and (E,F) energy expenditure in mice fed the indicated diets for approximately two months. (n = 5–7/group; statistics for the overall effects of diet, antibiotic (ABX) treatment, and the interaction represent the p-value from a two-way ANOVA; *p < 0.05 from a Sidak’s post-test examining the effect of parameters identified as significant in the two-way ANOVA). Error bars represent SEM.

References

    1. International Diabetes Federation. IDF Diabetes Atlas, 8th edn. Brussels, Belgium: International Diabetes Federation, (2017).
    1. Diabetes mellitus: a major risk factor for cardiovascular disease A joint editorial statement by the American Diabetes Association; The National Heart, Lung, and Blood Institute; The Juvenile Diabetes Foundation International; The National Institute of Diabetes and Digestive and Kidney Diseases; and The American Heart Association. Circulation. 1999;100:1132–1133. doi: 10.1161/01.CIR.100.10.1132.
    1. Giovannucci E, et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010;33:1674–1685. doi: 10.2337/dc10-0666.
    1. Barbagallo M, Dominguez LJ. Type 2 diabetes mellitus and Alzheimer’s disease. World J Diabetes. 2014;5:889–893. doi: 10.4239/wjd.v5.i6.889.
    1. Weickert MO. Nutritional modulation of insulin resistance. Scientifica. 2012;2012:424780. doi: 10.6064/2012/424780.
    1. Malik VS, Hu FB. Popular weight-loss diets: from evidence to practice. Nat Clin Pract Cardiovasc Med. 2007;4:34–41. doi: 10.1038/ncpcardio0726.
    1. Due A, Toubro S, Skov AR, Astrup A. Effect of normal-fat diets, either medium or high in protein, on body weight in overweight subjects: a randomised 1-year trial. Int J Obes Relat Metab Disord. 2004;28:1283–1290. doi: 10.1038/sj.ijo.0802767.
    1. Skov AR, Toubro S, Ronn B, Holm L, Astrup A. Randomized trial on protein vs carbohydrate in ad libitum fat reduced diet for the treatment of obesity. Int J Obes Relat Metab Disord. 1999;23:528–536. doi: 10.1038/sj.ijo.0800867.
    1. Weigle DS, et al. A high-protein diet induces sustained reductions in appetite, ad libitum caloric intake, and body weight despite compensatory changes in diurnal plasma leptin and ghrelin concentrations. Am J Clin Nutr. 2005;82:41–48. doi: 10.1093/ajcn/82.1.41.
    1. Campos-Nonato I, Hernandez L, Barquera S. Effect of a High-Protein Diet versus Standard-Protein Diet on Weight Loss and Biomarkers of Metabolic Syndrome: A Randomized Clinical Trial. Obes Facts. 2017;10:238–251. doi: 10.1159/000471485.
    1. Sluijs I, et al. Dietary intake of total, animal, and vegetable protein and risk of type 2 diabetes in the European Prospective Investigation into Cancer and Nutrition (EPIC)-NL study. Diabetes Care. 2010;33:43–48. doi: 10.2337/dc09-1321.
    1. Lagiou P, et al. Low carbohydrate-high protein diet and mortality in a cohort of Swedish women. J Intern Med. 2007;261:366–374. doi: 10.1111/j.1365-2796.2007.01774.x.
    1. Levine ME, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19:407–417. doi: 10.1016/j.cmet.2014.02.006.
    1. Solon-Biet SM, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19:418–430. doi: 10.1016/j.cmet.2014.02.009.
    1. Solon-Biet SM, et al. Dietary Protein to Carbohydrate Ratio and Caloric Restriction: Comparing Metabolic Outcomes in Mice. Cell reports. 2015;11:1529–1534. doi: 10.1016/j.celrep.2015.05.007.
    1. Mair W, Piper MD, Partridge L. Calories do not explain extension of life span by dietary restriction in Drosophila. PLoS Biol. 2005;3:e223. doi: 10.1371/journal.pbio.0030223.
    1. Lee KP, et al. Lifespan and reproduction in Drosophila: New insights from nutritional geometry. Proc Natl Acad Sci USA. 2008;105:2498–2503. doi: 10.1073/pnas.0710787105.
    1. Maida A, et al. A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution. J Clin Invest. 2016;126:3263–3278. doi: 10.1172/JCI85946.
    1. Cummings NE, et al. Restoration of metabolic health by decreased consumption of branched-chain amino acids. The Journal of physiology. 2018;596:623–645. doi: 10.1113/JP275075.
    1. Laeger T, et al. FGF21 is an endocrine signal of protein restriction. J Clin Invest. 2014;124:3913–3922. doi: 10.1172/JCI74915.
    1. Laeger T, et al. Metabolic Responses to Dietary Protein Restriction Require an Increase in FGF21 that Is Delayed by the Absence of GCN2. Cell reports. 2016;16:707–716. doi: 10.1016/j.celrep.2016.06.044.
    1. Fontana L, et al. Decreased Consumption of Branched-Chain Amino Acids Improves Metabolic Health. Cell reports. 2016;16:520–530. doi: 10.1016/j.celrep.2016.05.092.
    1. Keipert S, et al. Long-Term Cold Adaptation Does Not Require FGF21 or UCP1. Cell Metab. 2017;26:437–446 e435. doi: 10.1016/j.cmet.2017.07.016.
    1. Yu, D. et al. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J, fj201701211R, 10.1096/fj.201701211R (2018).
    1. Kreznar JH, et al. Host Genotype and Gut Microbiome Modulate Insulin Secretion and Diet-Induced Metabolic Phenotypes. Cell reports. 2017;18:1739–1750. doi: 10.1016/j.celrep.2017.01.062.
    1. Ridaura VK, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214. doi: 10.1126/science.1241214.
    1. Wu H, et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med. 2017;23:850–858. doi: 10.1038/nm.4345.
    1. Sung MM, et al. Improved Glucose Homeostasis in Obese Mice Treated With Resveratrol Is Associated With Alterations in the Gut Microbiome. Diabetes. 2017;66:418–425. doi: 10.2337/db16-0680.
    1. Rabot S, et al. High fat diet drives obesity regardless the composition of gut microbiota in mice. Scientific reports. 2016;6:32484. doi: 10.1038/srep32484.
    1. Greiner TU, Hyotylainen T, Knip M, Backhed F, Oresic M. The gut microbiota modulates glycaemic control and serum metabolite profiles in non-obese diabetic mice. PLoS One. 2014;9:e110359. doi: 10.1371/journal.pone.0110359.
    1. Vogt NM, et al. Gut microbiome alterations in Alzheimer’s disease. Scientific reports. 2017;7:13537. doi: 10.1038/s41598-017-13601-y.
    1. Turnbaugh PJ, et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1:6ra14. doi: 10.1126/scitranslmed.3000322.
    1. De Filippo C, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA. 2010;107:14691–14696. doi: 10.1073/pnas.1005963107.
    1. Xie C, et al. An Intestinal Farnesoid X Receptor-Ceramide Signaling Axis Modulates Hepatic Gluconeogenesis in Mice. Diabetes. 2017;66:613–626. doi: 10.2337/db16-0663.
    1. Sayin SI, et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 2013;17:225–235. doi: 10.1016/j.cmet.2013.01.003.
    1. Holmes AJ, et al. Diet-Microbiome Interactions in Health Are Controlled by Intestinal Nitrogen Source Constraints. Cell Metab. 2017;25:140–151. doi: 10.1016/j.cmet.2016.10.021.
    1. Zhu Y, et al. Meat, dairy and plant proteins alter bacterial composition of rat gut bacteria. Scientific reports. 2015;5:15220. doi: 10.1038/srep15220.
    1. Pluznick JL, et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA. 2013;110:4410–4415. doi: 10.1073/pnas.1215927110.
    1. Gul SS, et al. Inhibition of the gut enzyme intestinal alkaline phosphatase may explain how aspartame promotes glucose intolerance and obesity in mice. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme. 2017;42:77–83. doi: 10.1139/apnm-2016-0346.
    1. Krautkramer KA, et al. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol Cell. 2016;64:982–992. doi: 10.1016/j.molcel.2016.10.025.
    1. Mutel E, et al. Control of blood glucose in the absence of hepatic glucose production during prolonged fasting in mice: induction of renal and intestinal gluconeogenesis by glucagon. Diabetes. 2011;60:3121–3131. doi: 10.2337/db11-0571.
    1. Lamming DW, et al. Depletion of Rictor, an essential protein component of mTORC2, decreases male lifespan. Aging Cell. 2014;13:911–917. doi: 10.1111/acel.12256.
    1. Klindworth A, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41:e1. doi: 10.1093/nar/gks808.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nature methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Jiang H, Lei R, Ding SW, Zhu S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics. 2014;15:182. doi: 10.1186/1471-2105-15-182.
    1. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. 2011;27:2957–2963. doi: 10.1093/bioinformatics/btr507.
    1. DeSantis TZ, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol. 2006;72:5069–5072. doi: 10.1128/AEM.03006-05.
    1. Caporaso JG, et al. PyNAST: a flexible tool for aligning sequences to a template alignment. Bioinformatics. 2010;26:266–267. doi: 10.1093/bioinformatics/btp636.
    1. Paulson JN, Stine OC, Bravo HC, Pop M. Differential abundance analysis for microbial marker-gene surveys. Nature methods. 2013;10:1200–1202. doi: 10.1038/nmeth.2658.
    1. McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 2012;40:4288–4297. doi: 10.1093/nar/gks042.
    1. org.Mm.eg.db: Genome wide annotation for Mouse v. R package version 3.6.0 (2018).
    1. R: A language and environment for statistical computing (R Foundation for Statistical Computing, Vienna, Austria, 2018).
    1. Metsalu T, Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap. Nucleic Acids Res. 2015;43:W566–570. doi: 10.1093/nar/gkv468.
    1. Harputlugil E, et al. The TSC complex is required for the benefits of dietary protein restriction on stress resistance in vivo. Cell reports. 2014;8:1160–1170. doi: 10.1016/j.celrep.2014.07.018.
    1. Gong Q, et al. Fibroblast growth factor 21 improves hepatic insulin sensitivity by inhibiting mammalian target of rapamycin complex 1 in mice. Hepatology. 2016;64:425–438. doi: 10.1002/hep.28523.
    1. Utzschneider KM, Kratz M, Damman CJ, Hullar M. Mechanisms Linking the Gut Microbiome and Glucose Metabolism. J Clin Endocrinol Metab. 2016;101:1445–1454. doi: 10.1210/jc.2015-4251.
    1. Kuipers F, Bloks VW, Groen AK. Beyond intestinal soap–bile acids in metabolic control. Nature reviews. Endocrinology. 2014;10:488–498. doi: 10.1038/nrendo.2014.60.
    1. Fu T, et al. FXR Primes the Liver for Intestinal FGF15 Signaling by Transient Induction of beta-Klotho. Mol Endocrinol. 2016;30:92–103. doi: 10.1210/me.2015-1226.
    1. Jung D, et al. FXR agonists and FGF15 reduce fecal bile acid excretion in a mouse model of bile acid malabsorption. J Lipid Res. 2007;48:2693–2700. doi: 10.1194/jlr.M700351-JLR200.
    1. Tarling EJ, et al. RNA-binding protein ZFP36L1 maintains posttranscriptional regulation of bile acid metabolism. J Clin Invest. 2017;127:3741–3754. doi: 10.1172/JCI94029.
    1. Huang X, et al. Effects of dietary protein to carbohydrate balance on energy intake, fat storage, and heat production in mice. Obesity (Silver Spring) 2013;21:85–92. doi: 10.1002/oby.20007.
    1. Chakraborti CK. New-found link between microbiota and obesity. World J Gastrointest Pathophysiol. 2015;6:110–119. doi: 10.4291/wjgp.v6.i4.110.
    1. McEwen SA, Fedorka-Cray PJ. Antimicrobial use and resistance in animals. Clin Infect Dis. 2002;34(Suppl 3):S93–S106. doi: 10.1086/340246.
    1. Ozawa E. Studies on growth promotion by antibiotics. I. Effects of chlortetracycline on growth. J Antibiot (Tokyo) 1955;8:205–211.
    1. Cho I, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488:621–626. doi: 10.1038/nature11400.
    1. Butaye P, Devriese LA, Haesebrouck F. Antimicrobial growth promoters used in animal feed: effects of less well known antibiotics on gram-positive bacteria. Clin Microbiol Rev. 2003;16:175–188. doi: 10.1128/CMR.16.2.175-188.2003.
    1. Thevaranjan N, et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe. 2017;21:455–466 e454. doi: 10.1016/j.chom.2017.03.002.
    1. Spengler E, Loomba R. The Gut Microbiota, Intestinal Permeability, Bacterial Translocation, and Nonalcoholic Fatty Liver Disease: What Comes First? Cell Mol Gastroenterol Hepatol. 2015;1:129–130. doi: 10.1016/j.jcmgh.2015.01.007.
    1. Fontana L, Partridge L. Promoting health and longevity through diet: from model organisms to humans. Cell. 2015;161:106–118. doi: 10.1016/j.cell.2015.02.020.
    1. Cummings NE, Lamming DW. Regulation of metabolic health and aging by nutrient-sensitive signaling pathways. Molecular and cellular endocrinology. 2017;455:13–22. doi: 10.1016/j.mce.2016.11.014.
    1. Brown-Borg HM, Buffenstein R. Cutting back on the essentials: Can manipulating intake of specific amino acids modulate health and lifespan? Ageing Res Rev. 2017;39:87–95. doi: 10.1016/j.arr.2016.08.007.
    1. Lees EK, et al. Direct comparison of methionine restriction with leucine restriction on the metabolic health of C57BL/6J mice. Scientific reports. 2017;7:9977. doi: 10.1038/s41598-017-10381-3.
    1. Reimand J, et al. g:Profiler-a web server for functional interpretation of gene lists (2016 update) Nucleic Acids Res. 2016;44:W83–89. doi: 10.1093/nar/gkw199.

Source: PubMed

3
Sottoscrivi