Skip to content
1887

Abstract

Because large brains are energetically expensive, they are associated with metabolic traits that facilitate energy availability across vertebrates. However, the biological underpinnings driving these traits are not known. Given its role in regulating host metabolism in disease studies, we hypothesized that the gut microbiome contributes to variation in normal cross-vertebrate species differences in metabolism, including those associated with the brain’s energetic requirements. By inoculating germ-free mice with the gut microbiota (GM) of three primate species – two with relatively larger brains and one with a smaller brain – we demonstrated that the GM of larger-brained primates shifts host metabolism towards energy use and production, while that of smaller-brained primates stimulates energy storage in adipose tissues. Our findings establish a causal role of the GM in normal cross-host species differences in metabolism associated with relative brain size and suggest that the GM may have been an important facilitator of metabolic changes during human evolution that supported encephalization.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/mgen/10.1099/mgen.0.001322
2024-12-02
2025-04-20
Loading full text...

Full text loading...

/deliver/fulltext/mgen/10/12/mgen001322.html?itemId=/content/journal/mgen/10.1099/mgen.0.001322&mimeType=html&fmt=ahah

References

  1. Kuzawa CW, Chugani HT, Grossman LI, Lipovich L, Muzik O et al. Metabolic costs and evolutionary implications of human brain development. Proc Natl Acad Sci U S A 2014; 111:13010–13015 [View Article] [PubMed]
    [Google Scholar]
  2. Aiello LC, Wells JCK. Energetics and the evolution of the genus homo. Annu Rev Anthr 2002323–338 [View Article]
    [Google Scholar]
  3. Aiello LC, Wheeler P. The expensive-tissue hypothesis: the brain and the digestive system in human and primate evolution. Curr Anthropol 1995; 36:199–221 [View Article]
    [Google Scholar]
  4. Attwell D, Laughlin SB. An energy budget for signaling in the grey matter of the brain. J Cereb Blood Flow Metab 2001; 21:1133–1145 [View Article]
    [Google Scholar]
  5. Smaers JB, Rothman RS, Hudson DR, Balanoff AM, Beatty B et al. The evolution of mammalian brain size. Sci Adv 2021; 7:eabe2101 [View Article] [PubMed]
    [Google Scholar]
  6. Magden ER, Mansfield KG, Simmons JH, Abee CR. Nonhuman primates. In Fox JG, Anderson LC, Otto G, Pritchett-Corning KR, Whary MT. eds Laboratory Animal Medicine Cambridge, MA: Academic Press; 2015 pp 845–897 [View Article]
    [Google Scholar]
  7. Pontzer H, Brown MH, Raichlen DA, Dunsworth H, Hare B et al. Metabolic acceleration and the evolution of human brain size and life history. Nature 2016; 533:390–392 [View Article]
    [Google Scholar]
  8. Bauernfeind AL, Babbitt CC. Metabolic changes in human brain evolution. Evol Anthropol 2020; 29:201–211 [View Article]
    [Google Scholar]
  9. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vich Vila A et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 2019; 51:600–605 [View Article] [PubMed]
    [Google Scholar]
  10. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004; 101:15718–15723 [View Article] [PubMed]
    [Google Scholar]
  11. Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome 2019; 7:91 [View Article] [PubMed]
    [Google Scholar]
  12. Qin J, Li Y, Cai Z, Li S, Zhu J et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012; 490:55–60 [View Article]
    [Google Scholar]
  13. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013; 341:1241214 [View Article] [PubMed]
    [Google Scholar]
  14. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat Rev Gastroenterol Hepatol 2019; 16:461–478 [View Article]
    [Google Scholar]
  15. Lambert JE, Fellner V. In Vitro fermentation of dietary carbohydrates consumed by African apes and monkeys: preliminary results for interpreting microbial and digestive strategy. Int J Primatol 2012; 33:263–281 [View Article]
    [Google Scholar]
  16. Amato KR, G Sanders J, Song SJ, Nute M, Metcalf JL et al. Evolutionary trends in host physiology outweigh dietary niche in structuring primate gut microbiomes. ISME J 2019; 13:576–587 [View Article] [PubMed]
    [Google Scholar]
  17. Amato KR, Mallott EK, McDonald D, Dominy NJ, Goldberg T et al. Convergence of human and old world monkey gut microbiomes demonstrates the importance of human ecology over phylogeny. Genome Biol 2019; 20:201 [View Article] [PubMed]
    [Google Scholar]
  18. Leigh SR. Brain growth, life history, and cognition in primate and human evolution. Am J Primatol 2004; 62:139–164 [View Article] [PubMed]
    [Google Scholar]
  19. van Schaik CP, Triki Z, Bshary R, Heldstab SA. A farewell to the encephalization quotient: a new brain size measure for comparative primate cognition. Brain Behav Evol 2021; 96:1–12 [View Article] [PubMed]
    [Google Scholar]
  20. Navarrete A, van Schaik CP, Isler K. Energetics and the evolution of human brain size. Nature 2011; 480:91–93 [View Article]
    [Google Scholar]
  21. Cofran Z. Brain size growth in wild and captive chimpanzees (Pan troglodytes). Am J Primatol 2018; 80:e22876 [View Article] [PubMed]
    [Google Scholar]
  22. Arnoldini M, Sharma R, Moresi C, Chure G, Chabbey J et al. Quantifying the daily harvest of fermentation products from the human gut microbiota. Microbiology 2024 [View Article]
    [Google Scholar]
  23. McNeil NI. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr 1984; 39:338–342 [View Article] [PubMed]
    [Google Scholar]
  24. Conn AR, Fell DI, Steele RD. Characterization of alpha-keto acid transport across blood-brain barrier in rats. Am J Physiol 1983; 245:E253–60 [View Article] [PubMed]
    [Google Scholar]
  25. Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 2014; 5:1–11 [View Article]
    [Google Scholar]
  26. Wong JMW, de Souza R, Kendall CWC, Emam A, Jenkins DJA. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 2006; 40:235–243 [View Article] [PubMed]
    [Google Scholar]
  27. De Vadder F, Kovatcheva-Datchary P, Zitoun C, Duchampt A, Bäckhed F et al. Microbiota-produced succinate improves glucose homeostasis via intestinal gluconeogenesis. Cell Metab 2016; 24:151–157 [View Article]
    [Google Scholar]
  28. Purmal C, Kucejova B, Sherry AD, Burgess SC, Malloy CR et al. Propionate stimulates pyruvate oxidation in the presence of acetate. Am J Physiol Heart Circ Physiol 2014; 307:H1134–41 [View Article] [PubMed]
    [Google Scholar]
  29. Tirosh A, Calay ES, Tuncman G, Claiborn KC, Inouye KE et al. The short-chain fatty acid propionate increases glucagon and FABP4 production, impairing insulin action in mice and humans. Sci Transl Med 2019; 11:eaav0120 [View Article] [PubMed]
    [Google Scholar]
  30. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D et al. Acetate mediates a microbiome–brain–β-cell axis to promote metabolic syndrome. Nature 2016; 534:213–217 [View Article]
    [Google Scholar]
  31. Mackie RI, White BA. eds Gastrointestinal Microbiology New York: Chapman and Hall; 1997
    [Google Scholar]
  32. Leonard WR, Robertson ML. Nutritional requirements and human evolution: a bioenergetics model. Am J Hum Biol 1992; 4:179–195 [View Article] [PubMed]
    [Google Scholar]
  33. Vijay N, Morris ME. Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des 2014; 20:1487–1498 [View Article] [PubMed]
    [Google Scholar]
  34. Syamsunarno MRAA, Iso T, Hanaoka H, Yamaguchi A, Obokata M et al. A critical role of fatty acid binding protein 4 and 5 (FABP4/5) in the systemic response to fasting. PLoS One 2013; 8:e79386 [View Article] [PubMed]
    [Google Scholar]
  35. Clarke SD. Regulation of fatty acid synthase gene expression: an approach for reducing fat accumulation. J Anim Sci 1993; 71:1957–1965 [View Article] [PubMed]
    [Google Scholar]
  36. Lin X, Jia J, Du T, Li W, Wang X et al. Overexpression of miR-155 in the liver of transgenic mice alters the expression profiling of hepatic genes associated with lipid metabolism. PLoS One 2015; 10:e0118417 [View Article]
    [Google Scholar]
  37. Zhou Y, Rui L. Major urinary protein regulation of chemical communication and nutrient metabolism. Vitam Horm 2010; 83:151–163 [View Article] [PubMed]
    [Google Scholar]
  38. Burgess SC, He T, Yan Z, Lindner J, Sherry AD et al. Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver. Cell Metab 2007; 5:313–320 [View Article] [PubMed]
    [Google Scholar]
  39. Sunny NE, Parks EJ, Browning JD, Burgess SC. Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease. Cell Metab 2011; 14:804–810 [View Article] [PubMed]
    [Google Scholar]
  40. Leiter O, Walker TL. Platelets: the missing link between the blood and brain?. Prog Neurobiol 2019; 183:101695 [View Article] [PubMed]
    [Google Scholar]
  41. Burnouf T, Walker TL. The multifaceted role of platelets in mediating brain function. Blood 2022; 140:815–827 [View Article]
    [Google Scholar]
  42. Walker R, Hill K, Burger O, Hurtado AM. Life in the slow lane revisited: ontogenetic separation between chimpanzees and humans. Am J Phys Anthropol 2006; 129:577–583 [View Article]
    [Google Scholar]
  43. Aronoff JE, Ragin A, Wu C, Markl M, Schnell S et al. Why do humans undergo an adiposity rebound? Exploring links with the energetic costs of brain development in childhood using MRI-based 4D measures of total cerebral blood flow. Int J Obes 2022; 46:1044–1050 [View Article]
    [Google Scholar]
  44. Isler K, van Schaik CP. The expensive brain: a framework for explaining evolutionary changes in brain size. J Hum Evol 2009; 57:392–400 [View Article]
    [Google Scholar]
  45. Heldstab SA, Isler K, Graber SM, Schuppli C, van Schaik CP. The economics of brain size evolution in vertebrates. Curr Biol 2022; 32:R697–R708 [View Article] [PubMed]
    [Google Scholar]
  46. Huang Y, Mai CL, Liao WB, Kotrschal A. Body mass variation is negatively associated with brain size: evidence for the fat‐brain trade‐off in anurans. Evolution 2020; 74:1551–1557 [View Article]
    [Google Scholar]
  47. Kuzawa CW, Blair C. A hypothesis linking the energy demand of the brain to obesity risk. Proc Natl Acad Sci U S A 2019; 116:13266–13275 [View Article] [PubMed]
    [Google Scholar]
  48. Kuzawa CW. Adipose tissue in human infancy and childhood: An evolutionary perspective. Am J Phys Anthropol 1998; 107:177–209 [View Article]
    [Google Scholar]
  49. Cunnane SC, Crawford MA. Survival of the fattest: fat babies were the key to evolution of the large human brain. Comp Biochem Physiol A 2003; 136:17–26 [View Article]
    [Google Scholar]
  50. Leonard WR, Robertson ML, Snodgrass JJ. Energetics and the evolution of brain size in early homo. In Roebroeks W. eds Guts and Brains: An Integrative Approach to the Hominin Record Utrecht: Leiden University Press; 2007
    [Google Scholar]
  51. Jacobs B, Chugani HT, Allada V, Chen S, Phelps ME et al. Developmental changes in brain metabolism in sedated rhesus macaques and vervet monkeys revealed by positron emission tomography. Cereb Cortex 1995; 5:222–233 [View Article] [PubMed]
    [Google Scholar]
  52. Chugani HT, Phelps ME, Mazziotta JC. Positron emission tomography study of human brain functional development. Ann Neurol 1987; 22:487–497 [View Article] [PubMed]
    [Google Scholar]
  53. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH, Linden DR et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 2015; 29:1395–1403 [View Article] [PubMed]
    [Google Scholar]
  54. Yabut JM, Crane JD, Green AE, Keating DJ, Khan WI et al. Emerging roles for serotonin in regulating metabolism: new implications for an ancient molecule. Endocr Rev 2019; 40:1092–1107 [View Article] [PubMed]
    [Google Scholar]
  55. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013; 19:576–585 [View Article] [PubMed]
    [Google Scholar]
  56. Vuong HE, Pronovost GN, Williams DW, Coley EJL, Siegler EL et al. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020; 586:281–286 [View Article]
    [Google Scholar]
  57. Mallott EK, Amato KR. The microbial reproductive ecology of white-faced capuchins (Cebus capucinus). Am J Primatol 2018; 80:e22896 [View Article] [PubMed]
    [Google Scholar]
  58. Walters W, Hyde ER, Berg-Lyons D, Ackermann G, Humphrey G et al. Improved bacterial 16S rRNA gene (V4 and V4-5) and fungal internal transcribed spacer marker gene primers for microbial community surveys. mSystems 2016; 1:e00009–00015 [View Article]
    [Google Scholar]
  59. Nadkarni MA, Martin FE, Jacques NA, Hunter N. Determination of bacterial load by real-time PCR using a broad-range (universal) probe and primers set. Microbiology 2002; 148:257–266 [View Article] [PubMed]
    [Google Scholar]
  60. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet C et al. QIIME 2: reproducible, interactive, scalable, and extensible microbiome data science. Nat Biotech 2018; 37:852–857
    [Google Scholar]
  61. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J 2012; 6:610–618 [View Article] [PubMed]
    [Google Scholar]
  62. Willis A, Bunge J. Estimating diversity via frequency ratios. Biometrics 2015; 71:1042–1049 [View Article]
    [Google Scholar]
  63. Oksanen J. vegan: community ecology package. R. package; 2018
  64. Brooks ME, Kristensen K, van Benthem KJ, Magnusson A, Berg C et al. glmmTMB balances speed and flexibility among packages for zero-inflated generalized linear mixed modeling. R J 2017; 9:378 [View Article]
    [Google Scholar]
  65. Fox J, Weisberg S. An R Companion to Applied Regression Sage publications; 2018
    [Google Scholar]
  66. Klaus B, Strimmer K. Fdrtool: estimation of (local) false discovery rates and higher criticism. R package version 2015; 1:15
    [Google Scholar]
  67. Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018; 15:962–968 [View Article] [PubMed]
    [Google Scholar]
  68. Bates D, Maechler M, Bolker B, Walker S, Christensen RHB et al. Package ‘lme4. Convergence 2015; 12:
    [Google Scholar]
  69. Lohman BK, Weber JN, Bolnick DI. Evaluation of TagSeq, a reliable low-cost alternative for RNAseq. Mol Ecol Resour 2016; 16:1315–1321 [View Article] [PubMed]
    [Google Scholar]
  70. Meyer E, Aglyamova GV, Matz MV. Profiling gene expression responses of coral larvae (Acropora millepora) to elevated temperature and settlement inducers using a novel RNA-Seq procedure. Mol Ecol 2011; 20:3599–3616 [View Article] [PubMed]
    [Google Scholar]
  71. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J 2011; 17:10 [View Article]
    [Google Scholar]
  72. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9:357–359 [View Article] [PubMed]
    [Google Scholar]
  73. Love M. DESeq; 2014
  74. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B 1995; 57:289–300 [View Article]
    [Google Scholar]
  75. Reiman D, Layden BT, Dai Y. MiMeNet: exploring microbiome-metabolome relationships using neural networks. PLoS Comput Biol 2021; 17:e1009021 [View Article]
    [Google Scholar]
/content/journal/mgen/10.1099/mgen.0.001322
Loading
/content/journal/mgen/10.1099/mgen.0.001322
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error