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:

Neonatal exposure to a wild-derived microbiome protects mice against diet-induced obesity

Abstract

Obesity and its consequences are among the greatest challenges in healthcare. The gut microbiome is recognized as a key factor in the pathogenesis of obesity. Using a mouse model, we show here that a wild-derived microbiome protects against excessive weight gain, severe fatty liver disease and metabolic syndrome during a 10-week course of high-fat diet. This phenotype is transferable only during the first weeks of life. In adult mice, neither transfer nor severe disturbance of the wild-type microbiome modifies the metabolic response to a high-fat diet. The protective phenotype is associated with increased secretion of metabolic hormones and increased energy expenditure through activation of brown adipose tissue. Thus, we identify a microbiome that protects against weight gain and its negative consequences through metabolic programming in early life. Translation of these results to humans may identify early-life therapeutics that protect against obesity.

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: Wildlings are protected from excessive weight gain on HFD.
Fig. 2: Wildlings are protected from adverse metabolic effects of HFD.
Fig. 3: HFD-induced changes in the wildling and lab mouse microbiome.
Fig. 4: Impact of adult life microbiome on HFD response.
Fig. 5: Microbiota exposure in early-life programmes the response to HFD.
Fig. 6: Energy expenditure is increased in wildlings.
Fig. 7: Transfer of modified microbiome induces obesity resistance.

Similar content being viewed by others

Data availability

Raw sequence data from all 16S, shotgun metagenomics and RNA-sequencing experiments are deposited in the NCBI Sequence Read Archive under BioProject accession number PRJNA735448. Source data for the western blot are provided with this paper. Additional information and materials will be made available upon reasonable request.

Code availability

JAMS v.1.5.0 (https://github.com/johnmcculloch/JAMS_BW); virtual caloric chamber (https://sourceforge.net/projects/virtual-calorimeter/).

References

  1. WHO. The Global Health Observatory https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates/ghe-leading-causes-of-death (2020).

  2. Flegal, K. M., Kruszon-Moran, D., Carroll, M. D., Fryar, C. D. & Ogden, C. L. Trends in obesity among adults in the united states, 2005 to 2014. JAMA 315, 2284–2291 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Ogden, C. L. et al. Trends in obesity prevalence among children and adolescents in the united states, 1988–1994 through 2013–2014. JAMA 315, 2292–2299 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shreiner, A. B., Kao, J. Y. & Young, V. B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 31, 69–75 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Beura, L. K. et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 532, 512–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Clemente, J. C., Ursell, L. K., Parfrey, L. W. & Knight, R. The impact of the gut microbiota on human health: an integrative view. Cell 148, 1258–1270 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tilg, H., Zmora, N., Adolph, T. E. & Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 20, 40–54 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).

    Article  PubMed  Google Scholar 

  10. Kuang, Z. et al. The intestinal microbiota programs diurnal rhythms in host metabolism through histone deacetylase 3. Science 365, 1428–1434 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ridaura, V. K. et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341, 1241214 (2013).

    Article  PubMed  CAS  Google Scholar 

  12. Ferretti, P. et al. Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host Microbe 24, 133–145 e135 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Moeller, A. H. et al. Cospeciation of gut microbiota with hominids. Science 353, 380–382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sonnenburg, E. D. et al. Diet-induced extinctions in the gut microbiota compound over generations. Nature 529, 212–215 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Clemente, J. C. et al. The microbiome of uncontacted Amerindians. Sci. Adv. https://doi.org/10.1126/sciadv.1500183 (2015).

  16. Pasolli, E. et al. Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell 176, 649–662.e620 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sonnenburg, J. L. & Sonnenburg, E. D. Vulnerability of the industrialized microbiota. Science https://doi.org/10.1126/science.aaw9255 (2019).

  18. Collaborators, G. B. D. R. F. Global, regional, and national comparative risk assessment of 79 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388, 1659–1724 (2016).

    Article  Google Scholar 

  19. Blaser, M. J. The theory of disappearing microbiota and the epidemics of chronic diseases. Nat. Rev. Immunol. 17, 461–463 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Sonnenburg, E. D. & Sonnenburg, J. L. The ancestral and industrialized gut microbiota and implications for human health. Nat. Rev. Microbiol 17, 383–390 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Rosshart, S. P. et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science https://doi.org/10.1126/science.aaw4361 (2019).

  22. Rosshart, S. P. et al. Wild mouse gut microbiota promotes host fitness and improves disease resistance. Cell 171, 1015–1028.e1013 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chung, H. et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 149, 1578–1593 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Abdelaal, M., le Roux, C. W. & Docherty, N. G. Morbidity and mortality associated with obesity. Ann. Transl. Med. 5, 161 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Janochova, K., Haluzik, M. & Buzga, M. Visceral fat and insulin resistance—what we know? Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 163, 19–27 (2019).

    Article  PubMed  Google Scholar 

  26. Deng, F., Li, Y. & Zhao, J. The gut microbiome of healthy long-living people. Aging (Albany NY) 11, 289–290 (2019).

    Article  Google Scholar 

  27. Duvallet, C., Gibbons, S. M., Gurry, T., Irizarry, R. A. & Alm, E. J. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 8, 1784 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Bisanz, J. E., Upadhyay, V., Turnbaugh, J. A., Ly, K. & Turnbaugh, P. J. Meta-analysis reveals reproducible gut microbiome alterations in response to a high-fat diet. Cell Host Microbe 26, 265–272 e264 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Guo, J. & Hall, K. D. Estimating the continuous-time dynamics of energy and fat metabolism in mice. PLoS Comput. Biol. 5, e1000511 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Rui, L. Energy metabolism in the liver. Compr. Physiol. 4, 177–197 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  31. von Essen, G., Lindsund, E., Cannon, B. & Nedergaard, J. Adaptive facultative diet-induced thermogenesis in wild-type but not in UCP1-ablated mice. Am. J. Physiol. Endocrinol. Metab. 313, E515–E527 (2017).

    Article  CAS  Google Scholar 

  32. Zhang, Y. et al. Targeted deletion of thioesterase superfamily member 1 promotes energy expenditure and protects against obesity and insulin resistance. Proc. Natl Acad. Sci. USA 109, 5417–5422 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Boushey, R. P. & Drucker, D. J. Encyclopedia of Endocrine Disease (ed. Martini, L.) 183–188 (Elsevier, 2004) .

  34. Kirchner, H., Tong, J., Tschop, M. H. & Pfluger, P. T. Ghrelin and PYY in the regulation of energy balance and metabolism: lessons from mouse mutants. Am. J. Physiol. Endocrinol. Metab. 298, E909–E919 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Vrang, N., Madsen, A. N., Tang-Christensen, M., Hansen, G. & Larsen, P. J. PYY(3–36) reduces food intake and body weight and improves insulin sensitivity in rodent models of diet-induced obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 291, R367–R375 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Boey, D. et al. PYY transgenic mice are protected against diet-induced and genetic obesity. Neuropeptides 42, 19–30 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Breton, J. et al. Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab. 23, 324–334 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Kleinert, M., Sachs, S., Habegger, K. M., Hofmann, S. M. & Muller, T. D. Glucagon regulation of energy expenditure. Int. J. Mol. Sci. https://doi.org/10.3390/ijms20215407 (2019).

  39. Kinoshita, K. et al. Glucagon is essential for adaptive thermogenesis in brown adipose tissue. Endocrinology 155, 3484–3492 (2014).

    Article  PubMed  CAS  Google Scholar 

  40. Al Nabhani, Z. et al. A weaning reaction to microbiota is required for resistance to immunopathologies in the adult. Immunity 50, 1276–1288 e1275 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Cox, L. M. et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158, 705–721 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lynch, S. V. & Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 375, 2369–2379 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Negron, S. G., Ercan-Sencicek, A. G., Freed, J., Walters, M. & Lin, Z. Both proliferation and lipogenesis of brown adipocytes contribute to postnatal brown adipose tissue growth in mice. Sci. Rep. 10, 20335 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dominguez-Bello, M. G., Godoy-Vitorino, F., Knight, R. & Blaser, M. J. Role of the microbiome in human development. Gut 68, 1108–1114 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Ribo, S. et al. Increasing breast milk betaine modulates Akkermansia abundance in mammalian neonates and improves long-term metabolic health. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.abb0322 (2021).

  46. Wolf, M. J. et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S. & Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118, 229–241 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Evans, A. M. et al. High resolution mass spectrometry improves data quantity and quality as compared to unit mass resolution mass spectrometry in high-throughput profiling metabolomics. Metabolomics (Los Angel.) 2014, 2 (2014).

    Google Scholar 

  49. Ford, L. et al. Precision of a clinical metabolomics profiling platform for use in the identification of inborn errors of metabolism. J. Appl. Lab. Med. 5, 342–356 (2020).

    Article  PubMed  Google Scholar 

  50. Dehaven, C. D., Evans, A. M., Dai, H. & Lawton, K. A. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. J. Cheminform. 2, 9 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Evans, A. M., DeHaven, C. D., Barrett, T., Mitchell, M. & Milgram, E. Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems. Anal. Chem. 81, 6656–6667 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gower, J. C. Some distance properties of latent root and vector methods used in multivariate analysis. Biometrika 53, 325–338 (1966).

    Article  Google Scholar 

  55. Anderson, M. J. A new method for non-parametric multivariate analysis of variance. Austral. Ecol. 26, 32–46 (2001).

    Google Scholar 

  56. 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 

  57. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Li, D., Liu, C. M., Luo, R., Sadakane, K. & Lam, T. W. MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics 31, 1674–1676 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Seemann, T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 30, 2068–2069 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Wood, D. E. & Salzberg, S. L. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 15, R46 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Gu, S. et al. Mechanisms for complex chromosomal insertions. PLoS Genet. 12, e1006446 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Andrews, S. FastQC: A quality control tool for high throughput sequence data. http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (2010).

  64. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Ma, Mouse Metabolism Core, NIDDK, for assistance with 14C-DG uptake studies; M. Walters and Yuhai Dai, NIDDK Clinical Core, for assistance with the Mesoscale assay; C. Ohuigin, W. Yuan, G. Wallace and V. Thovarai (Cancer Inflammation Program Microbiome and Genetics Core, NCI) for 16S rRNA gene sequencing; B. Tran (Sequencing Facility at Leidos Biomedical Research, Inc./Frederick National Laboratory for Cancer Research) for sequencing; A. M. Cypress, R. Wess, J. Pydi (NIDDK) and R. Das Neves (NCI) for discussion; and the University of Michigan Animal Phenotyping Core for faecal bomb calorimetry. This study was funded by the intramural research programmes of the NIDDK and NCI, NIH; and by the NIH Director’s Challenge Award program and the DDIR Innovation Award program (B.R.). B.H. was supported by research fellowship no. HI 2088/1–1 from the Deutsche Forschungsgemeinschaft (DFG), Bonn, Germany.

Author information

Authors and Affiliations

Authors

Contributions

B.H. and B.R. conceived and designed the study. B.H., M.S.D., S.P.R. and B.R. designed the experiments. B.H. was responsible for the initial manuscript draft. B.H. and B.R. were responsible for the manuscript revision and editing. B.H., M.S.D., J.H.O., S.P.R. and O.G. performed the animal experiments. C.E.T. assisted with animal experiments. B.H., M.S.D. and J.H.B. analysed the 16S data. J.A.M. and G.T. analysed the metagenomic data. B.H. and R.U. analysed the transcriptomic data. J.G. and K.D.H. performed the virtual caloric chamber analysis. All authors discussed the data and commented on the manuscript.

Corresponding author

Correspondence to Barbara Rehermann.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary handling editor: George Caputa. Nature Metabolism thanks Gerard Eberl, Cathryn Nagler and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 HFD-induced changes in the cecal microbiome of lab and wildling mice.

Shotgun metagenomics data comparing the cecal microbiome of female wildling and lab mice at week 10 of chow or HFD. Lab mice on HFD: n = 5; lab mice on chow: n = 6; wildlings on HFD: n = 5; wildlings on chow: n = 5. a-c, Alpha-diversity, measured as number of observed unique taxa (a), Shannon index (b) and Simpson index (c) based on last-known taxa identified by shotgun metagenomics analysis of cecal microbiome of female wildling and lab mice at week 10 of chow diet or HFD. Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Unpaired two-tailed Student’s t test (Gaussian model) (a), Two-sided Wilcoxon signed rank test (b, c). (NS, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001). d, e, Heat maps generated by unsupervised clustering within lab (d) and wildling (e), showing the most variant last-known taxa (FDR-adjusted p < 0.05 by Mann Whitney Wilcoxon test) after filtering based on taxon genome completeness of >10% in at least 5% of samples and abundance of >250 parts per million in at least 15% of samples. Relative abundances are shown as z-scores, letters in front of last-known taxa describe taxonomy (s = species, f=family, g=genus, o=order, p=phylum).

Source data

Extended Data Fig. 2 Food consumption of wildling and lab mice on chow and HFD.

Data points represent daily kcal consumption per mouse, based on weekly measurements per cage over the course of chow or HFD diet. a, Data set refers to food consumption during experiments described in Fig. 1b; n = 55 lab mice on HFD, n = 42 lab mice on chow, n = 60 wildlings on HFD, n = 30 wildlings on chow. b, refers to Fig. 1d, n = 20 lab mice on HFD, n = 15 lab mice on chow, n = 23 wildlings on HFD, n = 17 wildlings on chow. c, refers to Fig. 4b, lab mice plus lab microbiota: n = 10 lab mice on chow, n = 24 lab mice on HFD; lab mice plus wildling microbiota: n = 10 mice on chow, n = 25 mice on HFD. d, refers to Fig. 4f; n = 10 lab mice, n = 10 wildlings. e, refers to Fig. 4j, n = 18 lab mice, n = 20 for wildlings. f, refers to Fig. 5c, n = 5 lab mice fostered by lab mice, n = 5 lab mice fostered by wildlings. g, refers to Fig. 5e, n = 8 lab mice fostered by lab mice, n = 8 lab mice fostered by wildlings. h, refers to Fig. 5h, n = 7 lab mice co-housed with lab mice, n = 10 lab mice co-housed with wildlings. i, refers to Fig. 5j, n = 13 lab mice co-housed with lab mice, n = 19 lab mice co-housed with wildlings. j, refers to Fig. 5m, n = 15 lab mice co-housed with lab mice, n = 10 lab mice co-housed with wildlings. k, refers to Fig. 5n, n = 15 lab mice co-housed with lab mice, n = 22 lab mice co-housed with wildlings. Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Unpaired two-tailed Student’s t test (Gaussian model) (c, d, f, g, h, i), two-sided Wilcoxon rank sum test (a, b, e, j, k). (NS, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, exact p-values are shown in the Source Data).

Source data

Extended Data Fig. 3 Fecal bomb calorimetry and rectal temperature of lab and wildling mice on chow and HFD.

a, b, Food energy intake (a) and fecal energy loss (bomb calorimetry) (b) in female lab or wildling mice on chow or HFD. N = 40 mice on chow and n = 40 mice on HFD examined per group over two independent experiments. c, Rectal temperature of male lab or wildling mice at week 9 of chow or HFD. N = 15 lab mice on chow, n = 20 lab mice on HFD, n = 17 wildlings on chow, n = 21 wildlings on HFD examined over two independent experiments. Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Two-sided Wilcoxon rank sum test (NS, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, exact p-values are shown in the Source Data).

Source data

Extended Data Fig. 4 Liver transcriptome of lab and wildling mice on HFD.

Bulk RNA sequencing of liver tissue from n = 7 lab mice (grey) and n = 7 wildlings (red) after 10 weeks of HFD. a, Unsupervised clustering of all metabolism-related gene transcripts that are significantly differently expressed. b, Volcano Plot. c, Comparison of lipogenesis-related gene transcripts (log2-fold expression) in liver tissue of the two groups of mice. Significance was determined as FDR ≤ 0.1.

Extended Data Fig. 5 Tissue-specific 2-deoxyglucose (2-DG) uptake in lab and wildling mice housed at 18 °C or 28 °C.

a, b, 2-DG uptake per milligram iBAT in female (a) or male (b) lab mice and wildlings at either 18 °C or 28 °C. c-h, 2-DG uptake in iWAT (c, d), pgWAT (e, f) or gastrocnemius muscle (g, h) in female (a, c, e, g) or male (b, d, f, h) lab or wildling mice held at 18 °C and 28 °C. N = 8 lab mice and n = 13 wildlings at 18 °C, n = 8 lab mice and n = 10 wildlings at 28 °C (a, c, e, g). N = 8 lab mice and n = 12 wildlings at 18 °C, n = 8 lab mice and n = 10 wildlings at 28 °C (b, d, f, h). Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. NS, not significant. Unpaired two-sided Student’s t test (Gaussian model) (a, b, d, e), two-sided Wilcoxon rank sum test (c, f, g, h). (NS, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, exact p-values are shown in the Source Data).

Source data

Extended Data Fig. 6 Western Blot analysis of AMPKalpha1, tyrosine hydroxylase and UCP1 in iBAT.

a, iBAT from lab and wildling mice at week 10 of HFD was analyzed by Western blot to quantitate the expression of tyrosine hydroxylase, AMPKalpha1 and UCP1 (upper bands) relative to GAPDH (lower bands). Brain lysate was used in the right lane (marked by asterisk). Representative Western Blot images for male mice (n = 6 mice per group). Unprocessed Western Blots are shown in Source Data. The experiment was independently repeated once for males and twice for females with similar results. b-g, The density of Western blot bands was quantified and normalized to each sample’s respective GAPDH band. Normalized expression levels are shown for tyrosine hydroxylase (b, c), AMPKalpha1 (d, e) and UCP1 (f, g) for female (b, d, f) and male (c, e, g) mice. N = 12 mice per group (b, c, e, g); n = 6 mice per group except n = 5 for the lab mouse group in blot 2 (d), n = 7 mice per group in blot 1, n = 6 mice per group in blot 2 (f). Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Two-sided Wilcoxon signed rank test (NS, not significant; ** p = 0.007538).

Source data

Extended Data Fig. 7 Metabolic hormones of lab and wildling mice on chow and HFD.

Serum concentration of metabolic hormones of lab mice and wildlings at baseline (week 10 of age on chow diet) and after 1 week and 10 weeks of HFD (weeks 11 and 20 of age, respectively). Each sample was pooled from two mice. Female lab mice at baseline, n = 14; at week 1 of HFD, n = 12; at week 10 of HFD, n = 16; male lab mice at baseline, n = 14; at week 1 of HFD, n = 14; at week 10 of HFD, n = 14; female wildlings at baseline, n = 14; at week 1 on HFD, n = 16; at week 10 of HFD, n = 12; male wildlings at baseline, n = 14; at week 1 of HFD, n = 16; at week 10 of HFD, n = 12. a Heatmap displaying median value of each group presented as z-score. BDNF, brain derived neurotropic factor; BAFF, B-cell activating factor. The Mesoscale Mouse Metabolic Combo 1 multiplex assay was used. b-h Serum concentration of glucagon (b), peptide YY (PYY) (c, d), thyroid stimulating hormone (TSH) (e, f), and the inactive form of glucagon like peptide 1 (GLP1) (g, h) at the indicated time points on chow or HFD as determined by multiplex Mesoscale Mouse Metabolic Combo 1 multiplex assay (b, c, d, g, h) or ELISA (e, f). Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Two-sided Wilcoxon signed rank test (NS, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, exact p-values are shown in the Source Data).

Source data

Extended Data Fig. 8 Increased diversity of wildling microbiome with interspersed clustering of MM-lab mice.

Shotgun metagenomics analysis of cecal microbiome from 12-day-old lab (n = 6), MM-lab (n = 4) and wildling (n = 5) pups. MM-lab mice were generated by co-housing lab mice with antibiotic-treated wildlings for 2 weeks and then separating and breeding these lab mice. Their offspring (microbiome modified (MM)-lab) were used for these experiments. a, Heat map generated by unsupervised clustering, showing the most variant last known taxa (LKT) after filtering based on taxon genome completeness of >10% in at least 5% of samples and abundance of >250 parts per million in at least 15% of samples. Relative abundances are shown as z-scores. Letters in front of LKT describe taxonomy (s = species, f=family, g=genus, o=order, p=phylum). b-d, Alpha-diversity, measured as number of observed unique taxa (b), Simpson index (c) and Shannon index (d) based on LKT. Box plots show median (center line), 75th (upper limit of box) and 25th percentile (lower limit of box) and outliers (whiskers) if values do not exceed 1.5-times interquartile range. Two-sided Wilcoxon signed rank test (NS, not significant; * p < 0.05, ** p < 0.01, exact p-values are shown in the Source Data).

Source data

Extended Data Fig. 9 Graphical Summary.

Exposure to a wildling microbiome resulting in increased brown adipose tissue activity that protects from diet-induced obesity later-on in life.

Supplementary information

Supplementary Information

Supplementary Table 1. Pathogen profile of wildling and lab mice.

Reporting Summary

Source data

Source Data Fig. 1

Exact P values for statistical results in Fig. 1.

Source Data Fig. 2

Exact P values for statistical results in Fig. 2.

Source Data Fig. 3

Exact P values for statistical results in Fig. 3.

Source Data Fig. 4

Exact P values for statistical results in Fig. 4.

Source Data Fig. 5

Exact P values for statistical results in Fig. 5.

Source Data Fig. 6

Exact P values for statistical results in Fig. 6.

Source Data Fig. 7

Exact P values for statistical results in Fig. 7.

Source Data Extended Data Fig. 1

Exact P values for statistical results in Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Exact P values for statistical results in Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Exact P values for statistical results in Extended Data Fig. 3.

Source Data Extended Data Fig. 5

Exact P values for statistical results in Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Unprocessed western blots for data shown in Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Exact P values for statistical results in Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Exact P values for statistical results in Extended Data Fig. 8.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hild, B., Dreier, M.S., Oh, J.H. et al. Neonatal exposure to a wild-derived microbiome protects mice against diet-induced obesity. Nat Metab 3, 1042–1057 (2021). https://doi.org/10.1038/s42255-021-00439-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00439-y

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