Gut microbiota plasticity is correlated with sustained weight loss on a low-carb or low-fat dietary intervention

Jessica A Grembi, Lan H Nguyen, Thomas D Haggerty, Christopher D Gardner, Susan P Holmes, Julie Parsonnet, Jessica A Grembi, Lan H Nguyen, Thomas D Haggerty, Christopher D Gardner, Susan P Holmes, Julie Parsonnet

Abstract

While low-carbohydrate and low-fat diets can both lead to weight-loss, a substantial variability in achieved long-term outcomes exists among obese but otherwise healthy adults. We examined the hypothesis that structural differences in the gut microbiota explain a portion of variability in weight-loss using two cohorts of obese adults enrolled in the Diet Intervention Examining The Factors Interacting with Treatment Success (DIETFITS) study. A total of 161 pre-diet fecal samples were sequenced from a discovery cohort (n = 66) and 106 from a validation cohort (n = 56). An additional 157 fecal samples were sequenced from the discovery cohort after 10 weeks of dietary intervention. We found no specific bacterial signatures associated with weight loss that were consistent across both cohorts. However, the gut microbiota plasticity (i.e. variability), was correlated with long-term (12-month) weight loss in a diet-dependent manner; on the low-fat diet subjects with higher pre-diet daily plasticity had higher sustained weight loss, whereas on the low-carbohydrate diet those with higher plasticity over 10 weeks of dieting had higher 12-month weight loss. Our findings suggest the potential importance of gut microbiota plasticity for sustained weight-loss. We highlight the advantages of evaluating kinetic trends and assessing reproducibility in studies of the gut microbiota.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Pre-diet microbial community composition not correlated with weight loss success. Pre-diet fecal microbiota collected from subjects in the (a) discovery and (b) validation cohorts prior to a low-carb (left) or low-fat (right) dietary intervention. Each point represents a single fecal sample and samples corresponding to the same subject are connected forming edges or triangles. Colors indicate 12-month weight loss success: very successful (VS), >10% weight loss; moderately successful (MS), 3–10% weight loss; and unsuccessful (US), <3% weight loss. The faded background polygons show convex hulls for corresponding success categories. Principal coordinates analysis was computed with Bray-Curtis distance on inverse-hyperbolic-sine transformed counts.
Figure 2
Figure 2
Gut microbiota plasticity over different periods. Pairwise β-diversity is shown between daily pre-diet samples (BL v. BL), between daily samples taken 10 weeks after initiation of the dietary intervention (10wk v. 10wk), and between BL and 10-week samples (BL v. 10wk). Bray-Curtis dissimilarities are shown for low-carb (left) and low-fat (right) diets. Grey points indicate computed pairwise dissimilarities between samples; colored points correspond to the average dissimilarity for each subject and are colored by weight loss category: US – Unsuccessful, <3% weight loss; MS – Moderately successful, 3–10% weight loss; VS – Very successful, >10% weight loss. Results with other β-diversity metrics are shown in Supplementary Figs. S2 and S3. The Wilcoxon rank sum test was performed to compare the mean difference in plasticity between US and VS groups.
Figure 3
Figure 3
Gut microbiota plasticity correlated with dietary change in a sex- and diet-dependent manner. Spearman’s rank correlations between (a) dietary change or (b) dietary adherence and plasticity (measured as Bray-Curtis dissimiliarity) between daily pre-diet samples (BL v. BL) and between pre-diet and 10-week samples (BL v. 10wk) are shown for low-carb (left) and low-fat (right) diets. Male (purple) and female (green) subjects show opposite correlations in many cases.
Figure 4
Figure 4
Differences in Prevotella/Bacteroides (P/B) ratio among weight loss success groups. Subjects on the low-carb diet are shown in the left panels; those on the low-fat diet are shown in the right panels. Grey points indicate P/B ratio for individual samples; colored points correspond to the average P/B ratio for each subject. Data from the (a) discovery and (b) validation cohort are displayed by subject’s weight loss success category at 12 months after the start of the dietary intervention: US – Unsuccessful; MS – Moderately successful; VS – Very successful. P-values shown for Wilcoxon rank-sum test comparing US and VS groups.
Figure 5
Figure 5
Taxa differentially abundant between weight loss success groups. ASV-clusters found differentially abundant when comparing subjects that were VS compared to US at 12-month weight loss on the low-carb diet. ASV-clusters were normalized and inverse-hyperbolic-sine-transformed for variance stabilization prior to analysis; the normalized, transformed values are shown on the y-axis. ASV-clusters have a median 96.8% sequence similarity (a taxonomic description can be found in Table 2). No taxa were found differentially abundant on the low-fat diet. Grey points represent individual samples and triangles represent the mean value for each subject. Triangle color represents the average log2-fold change (logFC) between subjects in the US vs VS groups; positive values indicate higher counts in VS subjects and negative values indicate higher counts in US subjects.

References

    1. Ng M, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. The Lancet. 2014;384:766–81. doi: 10.1016/S0140-6736(14)60460-8.
    1. Tremmel M, et al. Economic Burden of Obesity: A Systematic Literature Review. International Journal of Environmental Research and Public Health. 2017;14:435. doi: 10.3390/ijerph14040435.
    1. Johnston BC, et al. Comparison of weight loss among named diet programs in overweight and obese adults: A meta-analysis. JAMA - Journal of the American Medical Association. 2014;312:923–933. doi: 10.1001/jama.2014.10397.
    1. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the Atkins, Ornish, Weight Watchers, and Zone Diets for Weight Loss and Heart Disease Risk Reduction. JAMA. 2005;293:43. doi: 10.1001/jama.293.1.43.
    1. Alhassan, S., Kim, S., Bersamin, A., King, A. C. & Gardner, C. D. Dietary adherence and weight loss success among overweight women: Results from the A to Z weight loss study. International Journal of Obesity32, 985–991, 10.1038/ijo.2008.8, NIHMS150003 (2008).
    1. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2013;505:559–63. doi: 10.1038/nature12820.
    1. Walker AW, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. The ISME Journal. 2011;5:220–230. doi: 10.1038/ismej.2010.118.
    1. Ley R, Turnbaugh P, Klein S, Gordon J. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3. doi: 10.1038/nature4441021a.
    1. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature444, 1027–1031, 10.1038/nature05414, t8jd4qr3m (2006).
    1. Holzer, P., Reichmann, F. & Farzi, A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis, 10.1016/j.npep.2012.08.005 (2012).
    1. Cani, P. D. & Knauf, C. How gut microbes talk to organs: The role of endocrine and nervous routes, 10.1016/j.molmet.2016.05.011 (2016).
    1. Olivares, M. et al. The Potential Role of the Dipeptidyl Peptidase-4-Like Activity From the Gut Microbiota on the Host Health. Frontiers in Microbiology9, 10.3389/fmicb.2018.01900 (2018).
    1. Breton J, et al. Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metabolism. 2016;23:324–334. doi: 10.1016/j.cmet.2015.10.017.
    1. Lin, H. V. et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One7, 10.1371/journal.pone.0035240 (2012).
    1. Psichas A, et al. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. International Journal of Obesity. 2015;39:424–429. doi: 10.1038/ijo.2014.153.
    1. Healey GR, Murphy R, Brough L, Butts CA, Coad J. Interindividual variability in gut microbiota and host response to dietary interventions. Nutrition Reviews. 2017;75:1059–1080. doi: 10.1093/nutrit/nux062.
    1. Hjorth MF, et al. Pre-treatment microbial Prevotella-to-Bacteroides ratio, determines body fat loss success during a 6-month randomized controlled diet intervention. International Journal of Obesity. 2018;42:580–583. doi: 10.1038/ijo.2017.220.
    1. Hjorth MF, et al. Prevotella-to-Bacteroides ratio predicts body weight and fat loss success on 24-week diets varying in macronutrient composition and dietary fiber: results from a post-hoc analysis. International Journal of Obesity. 2019;43:149–157. doi: 10.1038/s41366-018-0093-2.
    1. Stanton M, et al. DIETFITS study (diet intervention examining the factors interacting with treatment success) – Study design and methods. Contemporary Clinical Trials. 2017;53:151–161. doi: 10.1016/j.cct.2016.12.021.
    1. Gardner CD, et al. Effect of Low-Fat vs Low-Carbohydrate Diet on 12-Month Weight Loss in Overweight Adults and the Association With Genotype Pattern or Insulin Secretion. JAMA. 2018;319:667–679. doi: 10.1001/jama.2018.0245.
    1. Wing RR, Phelan S. Long-term weight loss maintenance. Am. J. Clin. Nutr. 2005;82:222–227. doi: 10.1093/ajcn/82.1.222S.
    1. Jeffery RW, et al. Long-Term Maintenance of Weight Loss: Current Status. Heal. Psychol. 2000;19:5–16. doi: 10.1037/0278-6133.19.1(Suppl.).5.
    1. Costello EK, et al. Bacterial Community Variation in Human Body Habitats AcrossSpace and Time. Science. 2009;326:1694–1697. doi: 10.1126/science.1177486.
    1. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:4554–61. doi: 10.1073/pnas.1000087107.
    1. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–7. doi: 10.1038/nature11053.
    1. Flores, G. E. et al. Temporal variability is a personalized feature of the human microbiome. Genome biology15, 531, 10.1186/s13059-014-0531-y, NIHMS150003 (2014).
    1. Coyte KZ, Schluter J, Foster KR. The ecology of the microbiome: Networks, competition, and stability. Science (New York, N.Y.) 2015;350:663–6. doi: 10.1126/science.aad2602.
    1. Martínez I, et al. The Gut Microbiota of Rural Papua New Guineans: Composition, Diversity Patterns, and Ecological Processes. Cell Reports. 2015;11:527–538. doi: 10.1016/j.celrep.2015.03.049.
    1. Yachi S, Loreau M. Biodiversity and ecosystem productivity in a fluctuating environment: The insurance hypothesis. Proceedings of the National Academy of Sciences. 1999;96:1463–1468. doi: 10.1073/pnas.96.4.1463.
    1. Larraufie P, et al. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Scientific Reports. 2018;8:74. doi: 10.1038/s41598-017-18259-0.
    1. Byrne CS, Chambers ES, Morrison DJ, Frost G. The role of short chain fatty acids in appetite regulation and energy homeostasis. International Journal of Obesity. 2015;39:1331–1338. doi: 10.1038/ijo.2015.84.
    1. Gee JM, Johnson IT. Dietary lactitol fermentation increases circulating peptide YY and glucagon-like peptide-1 in rats and humans. Nutrition. 2005;21:1036–1043. doi: 10.1016/j.nut.2005.03.002.
    1. Cani PD, 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. American Journal of Clinical Nutrition. 2009;90:1236–1243. doi: 10.3945/ajcn.2009.28095.
    1. Batterham RL, et al. Gut hormone PYY3-36 physiologically inhibits food intake. Nature. 2002;418:650–654. doi: 10.1038/nature00887.
    1. Hirschberg AL. Sex hormones, appetite and eating behaviour in women. Maturitas. 2012;71:248–256. doi: 10.1016/j.maturitas.2011.12.016.
    1. Hirscbberg AL. Hormonal regulation of appetite and food intake. Annals Medicine. 1998;30:7–20. doi: 10.3109/07853899808999380.
    1. Hebert JR, et al. Gender Differences in Social Desirability and Social Approval Bias in Dietary Self-report. American Journal of Epidemiology. 1997;146:1046–1055. doi: 10.1093/oxfordjournals.aje.a009233.
    1. Faith JJ, Mcnulty NP, Rey FE, Gordon JI. Response to Diet in Gnotobiotic Mice. Science. 2011;333:101–105. doi: 10.1126/science.1206025.
    1. Shoaie S, et al. Quantifying diet-induced metabolic changes of the human gut microbiome. Cell Metabolism. 2015;22:320–331. doi: 10.1016/j.cmet.2015.07.001.
    1. Mcorist AL, et al. Fecal Butyrate Levels Vary Widely among Individuals but Are Usually Increased by a Diet High in Resistant Starch 1,2. J. Nutr. 2011;141:883–889. doi: 10.3945/jn.110.128504.
    1. Azad MB, et al. Infant gut microbiota and the hygiene hypothesis of allergic disease: impact of household pets and siblings on microbiota composition and diversity. Allergy, Asthma, & Clin. Immunol. 2013;9:1–9. doi: 10.1186/1710-1492-9-15.
    1. Song SJ, et al. Cohabiting family members share microbiota with one another and with their dogs. eLife. 2013;2013:458. doi: 10.7554/eLife.00458.
    1. Liang X, Bushman FD, FitzGerald GA. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Proc. Natl. Acad. Sci. 2015;112:10479–10484. doi: 10.1073/pnas.1501305112.
    1. Korpela, K. et al. Gut microbiota signatures predict host and microbiota responses to dietary interventions in obese individuals. PLoS One9, 10.1371/journal.pone.0090702 (2014).
    1. Dao MC, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut. 2016;65:426–436. doi: 10.1136/gutjnl-2014-308778.
    1. Harris PA, et al. Research electronic data capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. Journal of Biomedical Informatics. 2009;42:377–381. doi: 10.1016/j.jbi.2008.08.010.
    1. Hall KD, Kahan S. Maintenance of Lost Weight and Long-Term Management of Obesity. Medical Clinics of North America. 2018;102:183–197. doi: 10.1016/j.mcna.2017.08.012.
    1. Caporaso JG, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. The ISME J. 2012;6:1621–1624. doi: 10.1038/ismej.2012.8.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nature Methods. 2016;13:581–583. doi: 10.1038/nmeth.3869.
    1. Callahan, B., Sankaran, K., Fukuyama, J., McMurdie, P. & Holmes, S. Bioconductor workflow for microbiome data analysis: from raw reads to community analyses [version 1; referees: 3 approved]. F1000Research5, 10.12688/f1000research.8986.1 (2016).
    1. Wang, Q., Garrity, G. M., Tiedje, J. M. & Cole, J. R. Naïve Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73, 5261–5267, 10.1128/AEM.00062-07, Wang,Qiong,2007,Naive (2007).
    1. Quast C, et al. The silva ribosomal rna gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–D596. doi: 10.1093/nar/gks1219.
    1. Wright ES. Using decipher v2.0 to analyze big biological sequence data in r. The R J. 2016;8:352–359. doi: 10.32614/RJ-2016-025.
    1. Schliep K. phangorn: phylogenetic analysis in r. Bioinformatics. 2011;27:592–593. doi: 10.1093/bioinformatics/btq706.
    1. Bosshard PP, Abels S, Zbinden R, Böttger EC, Altwegg M. Ribosomal DNA Sequencing for Identification of Aerobic Gram-Positive Rods in the Clinical Laboratory (an 18-Month Evaluation) Journal of Clinical Microbiology. 2003;41:4134–4140. doi: 10.1128/JCM.41.9.4134-4140.2003.
    1. Větrovský T, Baldrian P. The variability of the 16s rrna gene in bacterial genomes and its consequences for bacterial community analyses. PLoS One. 2013;8:1–10. doi: 10.1371/journal.pone.0057923.
    1. Callahan BJ, McMurdie PJ, Holmes SP. Exact sequence variants should replace operational taxonomic units in marker-gene data analysis. ISME J. 2017;11:2639–2643. doi: 10.1038/ismej.2017.119.
    1. Nearing, J. T., Douglas, G. M., Comeau, A. M. & Langille, M. G. Denoising the Denoisers: An independent evaluation of microbiome sequence error-correction approaches.PeerJ2018, e5364, 10.7717/peerj.5364 (2018).
    1. Caruso, V., Song, X., Asquith, M. & Karstens, L. Performance of Microbiome Sequence Inference Methods in Environments with Varying Biomass. mSystems4, 10.1128/msystems.00163-18 (2019).
    1. Forster, D. et al. Improving eDNA-based protist diversity assessments using networks of amplicon sequence variants. Environ. Microbiol., 10.1111/1462-2920.14764 (2019).
    1. Oksanen, J. et al. vegan: Community Ecology Package, R package version 2.5-3 (2018).
    1. Nipperess DA, Matsen FA., IV The mean and variance of phylogenetic diversity under rarefaction. Methods Ecol Evol. 2013;4:566–572. doi: 10.1111/2041-210X.12042.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Laubscher NF. On stabilizing the binomial and negative binomial variances. J. Am. Stat. Assoc. 1961;56:143–150. doi: 10.1080/01621459.1961.10482100.
    1. Hoyle MH. Transformations: An introduction and a bibliography. Int. Stat. Rev./Revue Int. de Stat. 1973;41:203–223. doi: 10.2307/1402836.
    1. Burbidge JB, Magee L, Robb AL. Alternative transformations to handle extreme values of the dependent variable. J. Am. Stat. Assoc. 1988;83:123–127. doi: 10.1080/01621459.1988.10478575.
    1. Huber W, von Heydebreck A, Sültmann H, Poustka A, Vingron M. Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinformatics. 2002;18:S96–S104. doi: 10.1093/bioinformatics/18.suppl_1.S96.
    1. Benjamini, Y. & Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. Royal Stat. Soc. Ser. B (Methodological)57, 289–300, 10.2307/2346101, 95/57289 (1995).

Source: PubMed

3
Abonnere