Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Effects of underfeeding and oral vancomycin on gut microbiome and nutrient absorption in humans

Abstract

Direct evidence in humans for the impact of the microbiome on nutrient absorption is lacking. We conducted an extended inpatient study using two interventions that we hypothesized would alter the gut microbiome and nutrient absorption. In each, stool calorie loss, a direct proxy of nutrient absorption, was measured. The first phase was a randomized cross-over dietary intervention in which all participants underwent in random order 3 d of over- and underfeeding. The second was a randomized, double-blind, placebo-controlled pharmacologic intervention using oral vancomycin or matching placebo (NCT02037295). Twenty-seven volunteers (17 men and 10 women, age 35.1 ± 7.3, BMI 32.3 ± 8.0), who were healthy other than having impaired glucose tolerance and obesity, were enrolled and 25 completed the entire trial. The primary endpoints were the effects of dietary and pharmacological intervention on stool calorie loss. We hypothesized that stool calories expressed as percentage of caloric intake would increase with underfeeding compared with overfeeding and increase during oral vancomycin treatment. Both primary endpoints were met. Greater stool calorie loss was observed during underfeeding relative to overfeeding and during vancomycin treatment compared with placebo. Key secondary endpoints were to evaluate the changes in gut microbial community structure as evidenced by amplicon sequencing and metagenomics. We observed only a modest perturbation of gut microbial community structure with under- versus overfeeding but a more widespread change in community structure with reduced diversity with oral vancomycin. Increase in Akkermansia muciniphila was common to both interventions that resulted in greater stool calorie loss. These results indicate that nutrient absorption is sensitive to environmental perturbations and support the translational relevance of preclinical models demonstrating a possible causal role for the gut microbiome in dietary energy harvest.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Study outline.
Fig. 2: Dietary interventions impact nutrient absorption.
Fig. 3: Pharmacologic interventions impact nutrient absorption.
Fig. 4: Changes in gut microbiome in response to dietary interventions.
Fig. 5: Changes in the gut microbiome in response to oral vancomycin.
Fig. 6: Common changes in plasma butyrate and deoxycholic acid following dietary and pharmacologic interventions.

Similar content being viewed by others

Data availability

Data in the published article and its Supplementary Information have been presented where possible in aggregated form. The individual datasets generated during and/or analyzed during the current study are available from the corresponding author (A.B.) upon request, although restrictions may apply due to patient privacy and the General Data Protection Regulation. All sequencing data generated in the preparation of this manuscript have been deposited in NCBI’s Sequence Read Archive, with accession number PRJNA589622. Source data for Figs. 2–6 and Extended Data Figs. 2–8 are presented with the paper.

References

  1. Huang, K. C., Kormas, N., Steinbeck, K., Loughnan, G. & Caterson, I. D. Resting metabolic rate in severely obese diabetic and nondiabetic subjects. Obesity 12, 840–845 (2004).

    Article  Google Scholar 

  2. Ravussin, E. Energy metabolism in obesity: studies in the Pima Indians. Diabetes Care 16, 232–238 (1993).

    Article  CAS  PubMed  Google Scholar 

  3. Tataranni, P. et al. Body weight gain in free-living Pima Indians: effect of energy intake vs expenditure. Int. J. Obes. 27, 1578 (2003).

    Article  CAS  Google Scholar 

  4. Heymsfield, S. et al. Energy malabsorption: measurement and nutritional consequences. Am. J. Clin. Nutr. 34, 1954–1960 (1981).

    Article  CAS  PubMed  Google Scholar 

  5. Jumpertz, R. et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am. J. Clin. Nutr. 94, 58–65 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sommer, F. & Backhed, F. The gut microbiota—masters of host development and physiology. Nat. Rev. Microbiol. 11, 227–238 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Zmora, N., Suez, J. & Elinav, E. You are what you eat: diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 16, 35–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Blanton, L. V., Barratt, M. J., Charbonneau, M. R., Ahmed, T. & Gordon, J. I. Childhood undernutrition, the gut microbiota, and microbiota-directed therapeutics. Science 352, 1533 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Chagwedera, D. N. et al. Nutrient sensing in CD11c cells alters the gut microbiota to regulate food intake and body mass. Cell Metab. 30, 364–373.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Petersen, C. et al. T cell-mediated regulation of the microbiota protects against obesity. Science 365, eaat9351 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Allegretti, J. R. et al. Effects of fecal microbiota transplantation with oral capsules in obese patients. Clin. Gastroenterol. Hepatol. https://doi.org/10.1016/j.cgh.2019.07.006 (2019).

  13. Vrieze, A. et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143, 913–916.e7 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Block, J. P. et al. Early antibiotic exposure and weight outcomes in young children. Pediatrics 142, e20180290 (2018).

    Article  PubMed  Google Scholar 

  15. Isanaka, S. et al. Routine amoxicillin for uncomplicated severe acute malnutrition in children. N. Engl. J. Med. 374, 444–453 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Trehan, I. et al. Antibiotics as part of the management of severe acute malnutrition. N. Engl. J. Med. 368, 425–435 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Reijnders, D. et al. Effects of gut microbiota manipulation by antibiotics on host metabolism in obese humans: a randomized double-blind placebo-controlled trial. Cell Metab. 24, 63–74 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Murphy, E. F. et al. Divergent metabolic outcomes arising from targeted manipulation of the gut microbiota in diet-induced obesity. Gut 62, 220–226 (2013).

    Article  PubMed  Google Scholar 

  19. Yadav, H., Lee, J.-H., Lloyd, J., Walter, P. & Rane, S. G. Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. J. Biol. Chem. 288, 25088–25097 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Matzke, G., Zhanel, G. & Guay, D. Clinical pharmacokinetics of vancomycin. Clin Parmacokinet. 11, 257–282 (1986).

    Article  CAS  Google Scholar 

  21. Scheiman, J. et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat. Med. 25, 1104–1109 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tuohimaa, A., Riipinen, K. A., Brandt, K. & Alatossava, T. The genome of the virulent phage Lc-Nu of probiotic Lactobacillus rhamnosus, and comparative genomics with Lactobacillus casei phages. Arch. Virol. 151, 947–965 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Modi, S. R., Lee, H. H., Spina, C. S. & Collins, J. J. Antibiotic treatment expands the resistance reservoir and ecological network of the phage metagenome. Nature 499, 219–222 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Depommier, C. et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat. Med. 25, 1096–1103 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Reunanen, J. et al. Akkermansia muciniphila adheres to enterocytes and strengthens the integrity of the epithelial cell layer. Appl. Environ. Microbiol. 81, 3655–3662 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Everard, A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA 110, 9066–9071 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962–968 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wadden, D. et al. Circulating glucagon-like peptide-1 increases in response to short-term overfeeding in men. Nutr. Metab. 10, 33 (2013).

    Article  CAS  Google Scholar 

  29. Basolo, A. et al. Fasting glucagon-like peptide 1 concentration is associated with lower carbohydrate intake and increases with overeating. J. Endocrinol. Invest. 42, 557–566 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Kolaczynski, J. W., Ohannesian, J. P., Considine, R. V., Marco, C. C. & Caro, J. F. Response of leptin to short-term and prolonged overfeeding in humans. J. Clin. Endocrinol. Metab. 81, 4162–4165 (1996).

    CAS  PubMed  Google Scholar 

  31. Drucker, D. J. Glucagon-like peptide 2. J. Clin. Endocrinol. Metab. 86, 1759–1764 (2001).

    CAS  PubMed  Google Scholar 

  32. Guilmeau, S., Buyse, M. & Bado, A. Gastric leptin: a new manager of gastrointestinal function. Curr. Opin. Pharmacol. 4, 561–566 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Qin, X. et al. GLP-1 reduces intestinal lymph flow, triglyceride absorption, and apolipoprotein production in rats. Am. J. Physiol. Gastrointest. Liver Physiol. 288, G943–G949 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Liu, L. et al. Deoxycholic acid disrupts the intestinal mucosal barrier and promotes intestinal tumorigenesis. Food Funct. 9, 5588–5597 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Fasano, A. Intestinal permeability and its regulation by zonulin: diagnostic and therapeutic implications. Clin. Gastroenterol. Hepatol. 10, 1096–1100 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Guo, S., Al-Sadi, R., Said, H. M. & Ma, T. Y. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am. J. Pathol. 182, 375–387 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jones, M. L., Martoni, C. J., Ganopolsky, J. G., Labbé, A. & Prakash, S. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin. Biol. Ther. 14, 467–482 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Jumpertz, R. et al. Food label accuracy of common snack foods. Obesity 21, 164–169 (2013).

    Article  PubMed  Google Scholar 

  39. Allison, D. B., Heshka, S., Sepulveda, D. & Heymsfield, S. B. Counting calories—caveat emptor. JAMA 270, 1454–1456 (1993).

    Article  CAS  PubMed  Google Scholar 

  40. Urban, L. E. et al. The accuracy of stated energy contents of reduced-energy, commercially prepared foods. J. Am. Diet. Assoc. 110, 116–123 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ross, S. Functional foods: the Food and Drug Administration perspective. Am. J. Clin. Nutr. 71, 1735S–1738S (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Derrien, M., Vaughan, E. E., Plugge, C. M. & de Vos, W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 54, 1469–1476 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. König, J. et al. Human intestinal barrier function in health and disease. Clin. Transl. Gastroenterol. 7, e196 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Cani, P. D. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Depommier, C. et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat. Med. 25, 1096–1103 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Canani, R. B. et al. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J. Gastroenterol. 17, 1519 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Feng, Y., Wang, Y., Wang, P., Huang, Y. & Wang, F. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell. Physiol. Biochem. 49, 190–205 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Sapone, A. et al. Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes 55, 1443–1449 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Ohlsson, B., Orho-Melander, M. & Nilsson, P. M. Higher levels of serum zonulin may rather be associated with increased risk of obesity and hyperlipidemia, than with gastrointestinal symptoms or disease manifestations. Int. J. Mol. Sci. 18, 582 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  50. Munakata, A., Iwane, S., Todate, M., Nakaji, S. & Sugawara, K. Effects of dietary fiber on gastrointestinal transit time, fecal properties and fat absorption in rats. Tohoku J. Exp. Med. 176, 227–238 (1995).

    Article  CAS  PubMed  Google Scholar 

  51. Beyer, P. L. & Flynn, M. A. Effects of high-and low-fiber diets on human feces. J. Am. Diet. Assoc. 72, 271–277 (1978).

    Article  CAS  PubMed  Google Scholar 

  52. Hall, K. D. et al. Quantification of the effect of energy imbalance on bodyweight. Lancet 378, 826–837 (2011).

    Article  PubMed  Google Scholar 

  53. Ferraro, R., Boyce, V. L., Swinburn, B., De Gregorio, M. & Ravussin, E. Energy cost of physical activity on a metabolic ward in relationship to obesity. Am. J. Clin. Nutr. 53, 1368–1371 (1991).

    Article  CAS  PubMed  Google Scholar 

  54. Idiopathic, B. & Endocrinopathies, D. Report of the expert committee on the diagnosis and classification of diabetes mellitus. Diabetes Care 26, S5–S20 (2003).

    Article  Google Scholar 

  55. Zarling, E. J., Ruchim, M. A. & Makino, D. Improved technique for measuring fecal energy loss in normal and malabsorbing humans. J. Lab. Clin. Med. 107, 5–9 (1986).

    CAS  PubMed  Google Scholar 

  56. Thearle, M. S., Pannacciulli, N., Bonfiglio, S., Pacak, K. & Krakoff, J. Extent and determinants of thermogenic responses to 24 hours of fasting, energy balance, and five different overfeeding diets in humans. J. Clin. Endocrinol. Metab. 98, 2791–2799 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ravussin, E., Lillioja, S., Anderson, T. E., Christin, L. & Bogardus, C. Determinants of 24-hour energy expenditure in man. Methods and results using a respiratory chamber. J. Clin. Invest. 78, 1568–1578 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Gohl, D. M. et al. Systematic improvement of amplicon marker gene methods for increased accuracy in microbiome studies. Nat. Biotechnol. 34, 942 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, Q., Garrity, G. M., Tiedje, J. M. & Cole, J. R. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73, 5261–5267 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Dixon, P. VEGAN, a package of R functions for community ecology. J. Veg. Sci. 14, 927–930 (2003).

    Article  Google Scholar 

  63. Paradis, E., Claude, J. & Strimmer, K. APE: Analyses of Phylogenetics and Evolution in R language. Bioinformatics 20, 289–290 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Yu, G., Smith, D. K., Zhu, H., Guan, Y. & Lam, T. T. Y. ggtree: an R package for visualization and annotation of phylogenetic trees with their covariates and other associated data. Methods Ecol. Evol. 8, 28–36 (2017).

    Article  Google Scholar 

  66. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Segata, N. et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods 9, 811 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kanehisa, M., Sato, Y., Furumichi, M., Morishima, K. & Tanabe, M. New approach for understanding genome variations in KEGG. Nucleic Acids Res. 47, D590–D595 (2018).

    Article  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the volunteers who participated in the studies and the clinical staff of the Phoenix Epidemiology and Clinical Research Branch for conducting the examinations. This work was supported by the Intramural Research Program of the National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases. Additional support for the research in the Turnbaugh laboratory was provided by the National Institutes of Health (grant nos. R01HL122593, R21CA227232 and P30DK098722). The sponsors did not have any role in study design or analysis. P.J.T. holds an Investigators in the Pathogenesis of Infectious Disease Award from the Burroughs Wellcome Fund, is a Chan Zuckerberg Biohub investigator and is a Nadia’s Gift Foundation Innovator supported, in part, by the Damon Runyon Cancer Research Foundation (grant no. DRR-42-16) and the Searle Scholars Program (grant no. SSP-2016-1352). Q.Y.A. was the recipient of a graduate fellowship from A*STAR.

Author information

Authors and Affiliations

Authors

Contributions

J.K. and R.J.v.S. designed the study. A.B., M.H., S.H., P.P. and J.K. analyzed and interpreted the clinical data. M.W. and P.W. analyzed blood samples. S.P. assisted with laboratory procedures. Q.Y.A. and D.D.T. generated the microbiome data. Q.Y.A. led the data analysis. A.B. wrote the initial draft of the manuscript. J.K. and P.J.T. edited the submitted version. A.B., J.K. and P.J.T. are the guarantors of this work and have full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors critically revised the draft and approved the final manuscript.

Corresponding authors

Correspondence to Alessio Basolo, Peter J. Turnbaugh or Jonathan Krakoff.

Ethics declarations

Competing interests

P.J.T. is on the scientific advisory board for Kaleido, Pendulum, Seres and SNIPRbiome; there is no direct overlap between the current study and these consulting duties.

Additional information

Peer review information Joao Monteiro was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Consort diagram for study protocol.

Two individuals left the study due to personal reasons after completing the over and underfeeding portion of the study and after randomization to vancomycin (125 mg QID, 4 times per day) or placebo.

Extended Data Fig. 2 Urine calorie content in response to dietary and pharmaceutical interventions.

a, mean percent calorie loss in urine during overfeeding (n=25) and underfeeding (n=25). The Δ indicate the difference between underfeeding and overfeeding. Participants who started the dietary phase with overfeeding are represented by red dots, while those who started with underfeeding are represented by white dots. b, mean percent calorie loss in urine between vancomycin (n=11) and placebo (n=13) group. The sample size refers to the volunteers who had completed data of calorie loss from urine. p-values determined by two-sided paired (a) or two-sided unpaired Student’s t test (b).

Source data

Extended Data Fig. 3 Phylum-level relative abundances between overfeeding (OF) and underfeeding (UF).

Data points represent single samples (n=40 samples) and are colored by randomization group for the dietary phase where participants who started the dietary phase with overfeeding are represented by red dots (n=11), while those who started with underfeeding are represented by grey dots (n=9). Samples from the same individual are connected by a line. p-values were determined by two-sided paired Wilcoxon tests.

Source data

Extended Data Fig. 4 Effect of vancomycin treatment on gut microbiota composition.

Relative abundances of phyla significantly different between vancomycin and placebo groups (p<0.05, two-sided unpaired Wilcoxon test). Points indicate the mean value of samples from each subject (n=70 samples from 24 subjects). Boxplots indicate the inter-quartile range (IQR, 25th to 75th percentiles), with a center line indicating the median and whiskers showing the value ranges up to 1.5 x IQR above the 75th or below the 25th percentiles. b, Phylogenetic tree of 16S rRNA sequence variants (SVs) where colored tips denote significantly different SVs between vancomycin (n=11) and placebo (n=13) groups (FDR<0.1, DESeq2 with two-sided Wald test) with color representing fold change comparing vancomycin to placebo. Phylum distribution of the SVs are indicated around the tree.

Source data

Extended Data Fig. 5 Differentially abundant metabolic pathways between vancomycin and placebo.

Three metabolic pathways are differentially abundant between vancomycin (n=9) and placebo (n=10) groups (FDR-adjusted p-value, q<0.1, two-sided unpaired Wilcoxon test). Each point represents a single sample. Boxplots indicate the inter-quartile range (IQR, 25th to 75th percentiles), with a center line indicating the median and whiskers showing the value ranges up to 1.5 x IQR above the 75th or below the 25th percentiles.

Source data

Extended Data Fig. 6 Plasma acetate, propionate and lithocholic acid concentrations prior to and following treatment with oral vancomycin or placebo.

a, mean acetate concentrations (μM) during vancomycin (n=11) versus placebo (n=13) group. b, mean propionate concentrations (μM) during vancomycin (n=11) versus placebo (n=13) group. c, mean lithocholic acid concentrations (ng/ml) during vancomycin (n=4) versus placebo (n=11) group. The values are expressed as absolute values. The Δ indicate the difference (log expressed) between pre and post randomization measurements in vancomycin and placebo groups. p-values for the difference in acetate, propionate and lithocholic acid concentrations before and after randomization by vancomycin vs. placebo group as determined by two-sided unpaired Student’s t test. The sample size for each intervention refers to the volunteers who had available measurements of acetate, propionate and lithocholic acid concentrations.

Source data

Extended Data Fig. 7 Twenty-four-hour energy expenditure (24h-EE) and respiratory quotient (RQ) pre and post-randomization in vancomycin and placebo groups.

a, 24h-EE expressed as kcal/d; b, RQ, expressed as ratio; c, SMR expressed as kcal/d. d, SPA expressed as % of time spent moving over 24 hours. p-values indicate the change in 24h EE and its components (before and after randomization) between vancomycin (n=11) vs. placebo (n=11) groups as determined by two-sided Student’s unpaired t-test. The sample size for each intervention refers to the volunteers who had available measurements of 24h EE and its components. 24h EE: 24 hours energy expenditure; RQ: respiratory quotient; SMR: sleeping metabolic rate; SPA: spontaneous physical activity.

Source data

Extended Data Fig. 8 Glucose and insulin concentrations pre and post-randomization in vancomycin and placebo groups.

a, mean fasting glucose (mg/dL); b, mean 2-h glucose concentration (mg/dL). c, mean fasting insulin concentration (mU/L). d, mean 2-h insulin concentration (mU/L). p values indicate the change in glucose and insulin concentration (fasting and at 120’ during the OGTT) before and after randomization between vancomycin (n=12) vs. placebo (n=12) groups as determined by two-sided Student’s unpaired t-test. The sample size for each intervention refers to the volunteers who had available measurements of glucose and insulin during OGTT (oral glucose tolerance test). One volunteer in the vancomycin group did not have available measurement of fasting and 2-h insulin during OGTT.

Source data

Supplementary information

Supplementary Information

Consort checklist and protocols.

Reporting Summary

Supplementary Table

Supplementary Tables 1–12

Source data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Basolo, A., Hohenadel, M., Ang, Q.Y. et al. Effects of underfeeding and oral vancomycin on gut microbiome and nutrient absorption in humans. Nat Med 26, 589–598 (2020). https://doi.org/10.1038/s41591-020-0801-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0801-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing