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.

  • Review Article
  • Published:

Gut microbial metabolites as multi-kingdom intermediates

Abstract

The gut microbiota contributes to host physiology through the production of a myriad of metabolites. These metabolites exert their effects within the host as signalling molecules and substrates for metabolic reactions. Although the study of host–microbiota interactions remains challenging due to the high degree of crosstalk both within and between kingdoms, metabolite-focused research has identified multiple actionable microbial targets that are relevant for host health. Metabolites, as the functional output of combined host and microorganism interactions, provide a snapshot in time of an extraordinarily complex multi-organism system. Although substantial work remains towards understanding host–microbiota interactions and the underlying mechanisms, we review the current state of knowledge for each of the major classes of microbial metabolites with emphasis on clinical and translational research implications. We provide an overview of methodologies available for measurement of microbial metabolites, and in addition to discussion of key challenges, we provide a potential framework for integration of discovery-based metabolite studies with mechanistic work. Finally, we highlight examples in the literature where this approach has led to substantial progress in understanding host–microbiota interactions.

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: Fermentation of microbial-accessible carbohydrates and proteins by the colonic gut microbiota.
Fig. 2: Host–microbiota interactions during tryptophan metabolism.
Fig. 3: Gut microbiota–host interactions in one-carbon metabolism.

Similar content being viewed by others

References

  1. Sender, R., Fuchs, S. & Milo, R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 164, 337–340 (2016).

    CAS  PubMed  Google Scholar 

  2. Tierney, B. T. et al. The landscape of genetic content in the gut and oral human microbiome. Cell Host Microbe 26, 283–295.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Wikoff, W. R. et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl Acad. Sci. USA 106, 3698–3703 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Dodd, D. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 551, 648–652 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Martin, F.-P. J. et al. Panorganismal gut microbiome–host metabolic crosstalk. J. Proteome Res. 8, 2090–2105 (2009).

    CAS  PubMed  Google Scholar 

  6. Claus, S. P. et al. Systemic multicompartmental effects of the gut microbiome on mouse metabolic phenotypes. Mol. Syst. Biol. 4, 219 (2008).

    PubMed  PubMed Central  Google Scholar 

  7. Lupton, J. R. Microbial degradation products influence colon cancer risk: the butyrate controversy. J. Nutr. 134, 479–482 (2004).

    CAS  PubMed  Google Scholar 

  8. Donohoe, D. R. et al. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol. Cell 48, 612–626 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wahlström, A., Sayin, S. I., Marschall, H.-U. & Bäckhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 24, 41–50 (2016).

    PubMed  Google Scholar 

  10. Singh, V. et al. Dysregulated microbial fermentation of soluble fiber induces cholestatic liver cancer. Cell 175, 679–694.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Devlin, A. S. et al. Modulation of a circulating uremic solute via rational genetic manipulation of the gut microbiota. Cell Host Microbe 20, 709–715 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Deschasaux, M. et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat. Med. 24, 1526–1531 (2018).

    CAS  PubMed  Google Scholar 

  13. Martínez, I. et al. The gut microbiota of rural Papua New Guineans: composition, diversity patterns, and ecological processes. Cell Rep. 11, 527–538 (2015).

    PubMed  Google Scholar 

  14. Martínez, I., Kim, J., Duffy, P. R., Schlegel, V. L. & Walter, J. Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLoS ONE 5, e15046 (2010).

    PubMed  PubMed Central  Google Scholar 

  15. Jha, A. R. et al. Gut microbiome transition across a lifestyle gradient in Himalaya. PLoS Biol. 16, e2005396 (2018).

    PubMed  PubMed Central  Google Scholar 

  16. Vangay, P. et al. US immigration westernizes the human gut microbiome. Cell 175, 962–972.e10 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Smits, S. A. et al. Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 357, 802–806 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Davis, L. M. G., Martínez, I., Walter, J., Goin, C. & Hutkins, R. W. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS ONE 6, e25200 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Desai, M. S. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167, 1339–1353.e21 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Koh, A., De Vadder, F., Kovatcheva-Datchary, P. & Bäckhed, F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 165, 1332–1345 (2016).

    CAS  PubMed  Google Scholar 

  21. Macfarlane, S. & Macfarlane, G. T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 62, 67–72 (2007).

    Google Scholar 

  22. Makki, K., Deehan, E. C., Walter, J. & Bäckhed, F. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe 23, 705–715 (2018).

    CAS  PubMed  Google Scholar 

  23. Kaoutari, A. E., Armougom, F., Gordon, J. I., Raoult, D. & Henrissat, B. The abundance and variety of carbohydrate-active enzymes in the human gut microbiota. Nat. Rev. Microbiol. 11, 497–504 (2013).

    PubMed  Google Scholar 

  24. Bhattacharya, T., Ghosh, T. S. & Mande, S. S. Global profiling of carbohydrate active enzymes in human gut microbiome. PLoS ONE 10, e0142038 (2015).

    PubMed  PubMed Central  Google Scholar 

  25. McNeil, N. I. The contribution of the large intestine to energy supplies in man. Am. J. Clin. Nutr. 39, 338–342 (1984).

    CAS  PubMed  Google Scholar 

  26. Cummings, J. H., Pomare, E. W., Branch, W. J., Naylor, C. P. & Macfarlane, G. T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28, 1221–1227 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Besten, G. den et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, 2325–2340 (2013).

    Google Scholar 

  28. Smith, E. A. & Macfarlane, G. T. Dissimilatory amino acid metabolism in human colonic bacteria. Anaerobe 3, 327–337 (1997).

    CAS  PubMed  Google Scholar 

  29. Macfarlane, G. T., Gibson, G. R., Beatty, E. & Cummings, J. H. Estimation of short-chain fatty acid production from protein by human intestinal bacteria based on branched-chain fatty acid measurements. FEMS Microbiol. Lett. 101, 81–88 (1992).

    CAS  Google Scholar 

  30. Dalile, B., Oudenhove, L. V., Vervliet, B. & Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. 12, 453 (2019).

    Google Scholar 

  31. Johansson, M. E. V., Sjövall, H. & Hansson, G. C. The gastrointestinal mucus system in health and disease. Nat. Rev. Gastroenterol. 10, 352–361 (2013).

    CAS  Google Scholar 

  32. Schroeder, B. O. Fight them or feed them: how the intestinal mucus layer manages the gut microbiota. Gastroenterol. Rep. 7, 3–12 (2019).

    Google Scholar 

  33. Martens, E. C., Chiang, H. C. & Gordon, J. I. Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host Microbe 4, 447–457 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Sonnenburg, J. L. et al. Glycan foraging in vivo by an intestine-adapted bacterial symbiont. Science 307, 1955–1959 (2005).

    CAS  PubMed  Google Scholar 

  35. Cummings, J. H. & Macfarlane, G. T. The control and consequences of bacterial fermentation in the human colon. J. Appl. Bacteriol. 70, 443–459 (1991).

    CAS  PubMed  Google Scholar 

  36. Kukral, J. C., Adams, A. P. & Preston, F. W. Protein producing capacity of the human exocrine pancreas. Ann. Surg. 162, 63–73 (1965).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Silberberg, M. et al. The bioavailability of polyphenols is highly governed by the capacity of the intestine and of the liver to secrete conjugated metabolites. Eur. J. Nutr. 45, 88–96 (2006).

    CAS  PubMed  Google Scholar 

  38. Cardona, F., Andrés-Lacueva, C., Tulipani, S., Tinahones, F. J. & Queipo-Ortuño, M. I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr.Biochem. 24, 1415–1422 (2013).

    CAS  PubMed  Google Scholar 

  39. Puupponen-Pimiä, R. et al. Development of functional ingredients for gut health. Trends Food Sci. Tech. 13, 3–11 (2002).

    Google Scholar 

  40. Marín, L., Miguélez, E. M., Villar, C. J. & Lombó, F. Bioavailability of dietary polyphenols and gut microbiota metabolism: antimicrobial properties. Biomed. Res. Int. 2015, 1–18 (2015).

    Google Scholar 

  41. Roopchand, D. E. et al. Dietary polyphenols promote growth of the gut bacterium Akkermansia muciniphila and attenuate high-fat diet-induced metabolic syndrome. Diabetes 64, 2847–2858 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zeng, S.-L. et al. Citrus polymethoxyflavones attenuate metabolic syndrome by regulating gut microbiome and amino acid metabolism. Sci. Adv. 6, eaax6208 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ze, X., Duncan, S. H., Louis, P. & Flint, H. J. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J. 6, 1535–1543 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Deehan, E. C. et al. Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host Microbe 27, 389–404.e6 (2020).

    CAS  PubMed  Google Scholar 

  45. Bry, L., Falk, P. G., Midtvedt, T. & Gordon, J. I. A model of host–microbial interactions in an open mammalian ecosystem. Science 273, 1380–1383 (1996).

    CAS  PubMed  Google Scholar 

  46. David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2013).

    PubMed  PubMed Central  Google Scholar 

  47. Turnbaugh, P. J. et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 1, 6ra14 (2009).

    PubMed  PubMed Central  Google Scholar 

  48. Wu, G. D. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–108 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Clemente, J. C. et al. The microbiome of uncontacted Amerindians. Sci. Adv. 1, e1500183 (2015).

    PubMed  PubMed Central  Google Scholar 

  51. Holmes, A. J. et al. Diet–microbiome interactions in health are controlled by intestinal nitrogen source constraints. Cell Metab. 25, 140–151 (2017).

    CAS  PubMed  Google Scholar 

  52. Besten, G. den et al. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am. J. Physiol. Gastrointest. Liver Physiol. 305, G900–G910 (2013).

    Google Scholar 

  53. Reniere, M. L. Reduce, induce, thrive: bacterial redox sensing during pathogenesis. J. Bacteriol. 200, e00128-18 (2018).

    PubMed  PubMed Central  Google Scholar 

  54. Hylemon, P. B., Harris, S. C. & Ridlon, J. M. Metabolism of hydrogen gases and bile acids in the gut microbiome. FEBS Lett. 592, 2070–2082 (2018).

    CAS  PubMed  Google Scholar 

  55. Peng, L., Li, Z.-R., Green, R. S., Holzman, I. R. & Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 139, 1619–1625 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Lewis, K. et al. Enhanced translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflamm. Bowel Dis. 16, 1138–1148 (2010).

    PubMed  Google Scholar 

  57. Cherbut, C. et al. Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am. J. Physiol. Gastrointest. Liver Physiol. 275, G1415–G1422 (1998).

    CAS  Google Scholar 

  58. Soret, R. et al. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 138, 1772–1782.e4 (2010).

    CAS  PubMed  Google Scholar 

  59. Yano, J. M. et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161, 264–276 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Martin, A. M., Sun, E. W., Rogers, G. B. & Keating, D. J. The influence of the gut microbiome on host metabolism through the regulation of gut hormone release. Front. Physiol. 10, 428 (2019).

    PubMed  PubMed Central  Google Scholar 

  61. Fukumoto, S. et al. Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 284, R1269–R1276 (2003).

    CAS  PubMed  Google Scholar 

  62. Krautkramer, K. A. et al. Diet–microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol. Cell 64, 982–992 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Kaiko, G. E. et al. The colonic crypt protects stem cells from microbiota-derived metabolites. Cell 165, 1708–1720 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Donohoe, D. R. et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 4, 1387–1397 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Donohoe, D. R. et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 13, 517–526 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Vadder, F. D. et al. Microbiota-generated metabolites promote metabolic benefits via gut–brain neural circuits. Cell 156, 84–96 (2014).

    PubMed  Google Scholar 

  67. Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).

    CAS  PubMed  Google Scholar 

  68. Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8+ T cells. Immunity 51, 285–297.e5 (2019).

    CAS  PubMed  Google Scholar 

  69. Kasahara, K. et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 474, 327 (2018).

    Google Scholar 

  70. Samuel, B. S. et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl Acad. Sci. USA 105, 16767–16772 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  72. Canfora, E. E., Meex, R. C. R., Venema, K. & Blaak, E. E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 15, 261–273 (2019).

    CAS  PubMed  Google Scholar 

  73. Louis, P., Hold, G. L. & Flint, H. J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 12, 661–672 (2014).

    CAS  PubMed  Google Scholar 

  74. Husted, A. S., Trauelsen, M., Rudenko, O., Hjorth, S. A. & Schwartz, T. W. GPCR-mediated signaling of metabolites. Cell Metab. 25, 777–796 (2017).

    CAS  PubMed  Google Scholar 

  75. Newgard, C. B. et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 9, 311–326 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Newgard, C. B. Interplay between lipids and branched-chain amino acids in development of insulin resistance. Cell Metab. 15, 606–614 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Pedersen, H. K. et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376–381 (2016).

    CAS  PubMed  Google Scholar 

  78. Ericksen, R. E. et al. Loss of BCAA catabolism during carcinogenesis enhances mTORC1 activity and promotes tumor development and progression. Cell Metab. 29, 1151–1165.e6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. VanDusseldorp, T. A. et al. Effect of branched-chain amino acid supplementation on recovery following acute eccentric exercise. Nutrients 10, 1389 (2018).

    PubMed Central  Google Scholar 

  80. Evenepoel, P. et al. Amount and fate of egg protein escaping assimilation in the small intestine of humans. Am. J. Physiol. Gastrointest. Liver Physiol. 277, G935–G943 (1999).

    CAS  Google Scholar 

  81. Mahé, S., Huneau, J. F., Marteau, P., Thuillier, F. & Tomé, D. Gastroileal nitrogen and electrolyte movements after bovine milk ingestion in humans. Am. J. Clin. Nutr. 56, 410–416 (1992).

    PubMed  Google Scholar 

  82. Neis, E. P. J. G., Dejong, C. H. C. & Rensen, S. S. The role of microbial amino acid metabolism in host metabolism. Nutrients 7, 2930–2946 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Ratzke, C. & Gore, J. Modifying and reacting to the environmental pH can drive bacterial interactions. PLoS Biol. 16, e2004248 (2018).

    PubMed  PubMed Central  Google Scholar 

  84. Smith, E. A. & Macfarlane, G. T. Enumeration of human colonic bacteria producing phenolic and indolic compounds: effects of pH, carbohydrate availability and retention time on dissimilatory aromatic amino acid metabolism. J. Appl. Bacteriol. 81, 288–302 (1996).

    CAS  PubMed  Google Scholar 

  85. Birkett, A., Muir, J., Phillips, J., Jones, G. & O’Dea, K. Resistant starch lowers fecal concentrations of ammonia and phenols in humans. Am. J. Clin. Nutr. 63, 766–772 (1996).

    CAS  PubMed  Google Scholar 

  86. Stephen, A. M. & Cummings, J. H. Mechanism of action of dietary fibre in the human colon. Nature 284, 283–284 (1980).

    CAS  PubMed  Google Scholar 

  87. Sridharan, G. V. et al. Prediction and quantification of bioactive microbiota metabolites in the mouse gut. Nat. Commun. 5, 5492 (2014).

    CAS  PubMed  Google Scholar 

  88. Alkhalaf, L. M. & Ryan, K. S. Biosynthetic manipulation of tryptophan in bacteria: pathways and mechanisms. Chem. Biol. 22, 317–328 (2015).

    CAS  PubMed  Google Scholar 

  89. Agus, A., Planchais, J. & Sokol, H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23, 716–724 (2018).

    CAS  PubMed  Google Scholar 

  90. Cervenka, I., Agudelo, L. Z. & Ruas, J. L. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science 357, eaaf9794 (2017).

    PubMed  Google Scholar 

  91. Houtkooper, R. H., Cantó, C., Wanders, R. J. & Auwerx, J. The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. Endocr. Rev. 31, 194–223 (2010).

    CAS  PubMed  Google Scholar 

  92. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2010).

    PubMed  PubMed Central  Google Scholar 

  93. Rhee, S. J., Walker, W. A. & Cherayil, B. J. Developmentally regulated intestinal expression of IFN-γ and its target genes and the age-specific response to enteric Salmonella infection. J. Immunol. 175, 1127–1136 (2005).

    CAS  PubMed  Google Scholar 

  94. Zelante, T. et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).

    CAS  PubMed  Google Scholar 

  95. Vujkovic-Cvijin, I. et al. Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci. Transl. Med. 5, 193ra91–193ra91 (2013).

    PubMed  PubMed Central  Google Scholar 

  96. Agudelo, L. Z. et al. Skeletal muscle PGC-1α1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell 159, 33–45 (2014).

    CAS  PubMed  Google Scholar 

  97. Hubbard, T. D. et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci. Rep. 5, 12689 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Walther, D. J. et al. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science 299, 76–76 (2003).

    CAS  PubMed  Google Scholar 

  99. Mawe, G. M. & Hoffman, J. M. Serotonin signalling in the gut — functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. 10, 473–486 (2013).

    CAS  Google Scholar 

  100. Reigstad, C. S. et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 29, 1395–1403 (2014).

    PubMed  PubMed Central  Google Scholar 

  101. Valles-Colomer, M. et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 4, 623–632 (2019).

    CAS  PubMed  Google Scholar 

  102. Lukić, I. et al. Antidepressants affect gut microbiota and Ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior. Transl. Psychiat 9, 133 (2019).

    Google Scholar 

  103. Fung, T. C. et al. Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut. Nat. Microbiol. 4, 2064–2073 (2019).

    PubMed  PubMed Central  Google Scholar 

  104. Bhattarai, Y. et al. Gut microbiota-produced tryptamine activates an epithelial G-protein-coupled receptor to increase colonic secretion. Cell Host Microbe 23, 775–785.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Barcik, W., Wawrzyniak, M., Akdis, C. A. & O’Mahony, L. Immune regulation by histamine and histamine-secreting bacteria. Curr. Opin. Immunol. 48, 108–113 (2017).

    CAS  PubMed  Google Scholar 

  106. Koh, A. et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell 175, 947–961.e17 (2018).

    CAS  PubMed  Google Scholar 

  107. Rekdal, V. M., Bess, E. N., Bisanz, J. E., Turnbaugh, P. J. & Balskus, E. P. Discovery and inhibition of an interspecies gut bacterial pathway for levodopa metabolism. Science 364, eaau6323 (2019).

    PubMed Central  Google Scholar 

  108. Kessel, S. P. van et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat. Commun. 10, 310 (2019).

    PubMed  PubMed Central  Google Scholar 

  109. Nallu, A., Sharma, S., Ramezani, A., Muralidharan, J. & Raj, D. Gut microbiome in chronic kidney disease: challenges and opportunities. Transl. Res. 179, 24–37 (2017).

    CAS  PubMed  Google Scholar 

  110. Vanholder, R., Schepers, E., Pletinck, A., Nagler, E. V. & Glorieux, G. The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: a systematic review. J. Am. Soc. Nephrol. 25, 1897–1907 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Hatch, M. & Vaziri, N. D. Enhanced enteric excretion of urate in rats with chronic renal failure. Clin. Sci. 86, 511–516 (1994).

    CAS  Google Scholar 

  112. Einheber, A. & Carter, D. The role of the microbial flora in uremia. J. Exp. Med. 123, 239–250 (1966).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Werder, A. A., Amos, M. A., Nielsen, A. H. & Wolfe, G. H. Comparative effects of germfree and ambient environments on the development of cystic kidney disease in CFWwd mice. J. Lab. Clin. Med. 103, 399–407 (1984).

    CAS  PubMed  Google Scholar 

  114. Hallman, T. M. et al. The mitochondrial and kidney disease phenotypes of kd/kd mice under germfree conditions. J. Autoimmun. 26, 1–6 (2006).

    CAS  PubMed  Google Scholar 

  115. Consortium, C. K. D. P. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 375, 2073–2081 (2010).

    Google Scholar 

  116. Poesen, R. et al. Microbiota-derived phenylacetylglutamine associates with overall mortality and cardiovascular disease in patients with CKD. J. Am. Soc. Nephrol. 27, 3479–3487 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Nemet, I. et al. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell 180, 862–877.e22 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. de Aguiar Vallim, T. Q., Tarling, E. J. & Edwards, P. A. Pleiotropic roles of bile acids in metabolism. Cell Metab. 17, 657–669 (2013).

    PubMed  PubMed Central  Google Scholar 

  119. Sayin, S. I. et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-β-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 17, 225–235 (2013).

    CAS  PubMed  Google Scholar 

  120. Quinn, R. A. et al. Global chemical effects of the microbiome include new bile-acid conjugations. Nature 579, 123–129 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Funabashi, M. et al. A metabolic pathway for bile acid dehydroxylation by the gut microbiome. Nature 582, 566–570 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Schramm, C. Bile acids, the microbiome, immunity, and liver tumors. N. Engl. J. Med. 379, 888–890 (2018).

    PubMed  Google Scholar 

  123. Jia, W., Xie, G. & Jia, W. Bile acid–microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat. Rev. Gastroenterol. Hepatol. 15, 111–128 (2017).

    PubMed  PubMed Central  Google Scholar 

  124. Lund, M. L. et al. L-cell differentiation is induced by bile acids through GPBAR1 and paracrine GLP-1 and serotonin signaling. Diabetes 69, 614–623 (2020).

    CAS  PubMed  Google Scholar 

  125. Inagaki, T. et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2, 217–225 (2005).

    CAS  PubMed  Google Scholar 

  126. Ryan, K. K. et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature 509, 183–188 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Flynn, C. R. et al. Bile diversion to the distal small intestine has comparable metabolic benefits to bariatric surgery. Nat. Commun. 6, 7715 (2015).

    CAS  PubMed  Google Scholar 

  128. Schmitt, J. et al. Protective effects of farnesoid X receptor (FXR) on hepatic lipid accumulation are mediated by hepatic FXR and independent of intestinal FGF15 signal. Liver Int. 35, 1133–1144 (2014).

    PubMed  PubMed Central  Google Scholar 

  129. Jiang, C. et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat. Commun. 6, 10166 (2015).

    CAS  PubMed  Google Scholar 

  130. Li, F. et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat. Commun. 4, 2384 (2013).

    PubMed  Google Scholar 

  131. Sjöström, L. et al. Effects of bariatric surgery on cancer incidence in obese patients in Sweden (Swedish Obese Subjects Study): a prospective, controlled intervention trial. Lancet Oncol. 10, 653–662 (2009).

    PubMed  Google Scholar 

  132. Sjöström, L. et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N. Engl. J. Med. 351, 2683–2693 (2004).

    PubMed  Google Scholar 

  133. Tremaroli, V., Karlsson, F. & Werling, M. et al. Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation. Cell Metab. 22, 228–238 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Gottardi, A. D. et al. The bile acid nuclear receptor FXR and the bile acid binding protein IBABP are differently expressed in colon cancer. Dig. Dis. Sci. 49, 982–989 (2004).

    PubMed  Google Scholar 

  135. Yoshimoto, S. et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499, 97–101 (2013).

    CAS  PubMed  Google Scholar 

  136. Gadaleta, R. M. et al. Activation of bile salt nuclear receptor FXR is repressed by pro-inflammatory cytokines activating NF-κB signaling in the intestine. Biochim. Biophys. Acta 1812, 851–858 (2011).

    CAS  PubMed  Google Scholar 

  137. Bailey, A. M. et al. FXR silencing in human colon cancer by DNA methylation and KRAS signaling. Am. J. Physiol. Gastrointest. Liver Physiol. 306, G48–G58 (2014).

    CAS  PubMed  Google Scholar 

  138. Degirolamo, C. et al. Prevention of spontaneous hepatocarcinogenesis in farnesoid X receptor-null mice by intestinal-specific farnesoid X receptor reactivation. Hepatology 61, 161–170 (2014).

    PubMed  Google Scholar 

  139. Yang, F. et al. Spontaneous development of liver tumors in the absence of the bile acid receptor farnesoid X receptor. Cancer Res. 67, 863–867 (2007).

    CAS  PubMed  Google Scholar 

  140. Ma, C. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 360, eaan5931 (2018).

    PubMed  PubMed Central  Google Scholar 

  141. Ducker, G. S. & Rabinowitz, J. D. One-carbon metabolism in health and disease. Cell Metab. 25, 27–42 (2016).

    PubMed  PubMed Central  Google Scholar 

  142. Zeisel, S. H. & Costa, K. D. Choline: an essential nutrient for public health. Nutr. Rev. 67, 615–623 (2009).

    PubMed  Google Scholar 

  143. Craciun, S. & Balskus, E. P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl Acad. Sci. USA 109, 21307–21312 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Wang, Z. et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472, 57–63 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhu, W. et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165, 111–124 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Tang, W. H. W. et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 368, 1575–1584 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Li, X. S. et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 38, 814–824 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Seldin, M. M. et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J. Am. Heart Assoc. 5, e002767 (2016).

    PubMed  PubMed Central  Google Scholar 

  149. Koeth, R. A. et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Invest. 129, 373–387 (2018).

    PubMed  PubMed Central  Google Scholar 

  150. Li, X. S. et al. Trimethyllysine, a trimethylamine N-oxide precursor, provides near- and long-term prognostic value in patients presenting with acute coronary syndromes. Eur. Heart J. 40, 2700–2709 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Hill, M. J. Intestinal flora and endogenous vitamin synthesis. Eur. J. Cancer Prev. 6 (Suppl 1), S43–S45 (1997).

    PubMed  Google Scholar 

  152. Magnúsdóttir, S., Ravcheev, D., Crécy-Lagard, V. de & Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front. Genet. 6, 1–18 (2015).

    Google Scholar 

  153. Aufreiter, S. et al. Folate is absorbed across the colon of adults: evidence from cecal infusion of 13C-labeled [6S]-5-formyltetrahydrofolic acid. Am. J. Clin. Nutr. 90, 116–123 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Yatsunenko, T. et al. Human gut microbiome viewed across age and geography. Nature 486, 222–227 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Roberts, A. B. et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat. Med. 24, 1407–1417 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Zimmermann, M., Zimmermann-Kogadeeva, M., Wegmann, R. & Goodman, A. L. Separating host and microbiome contributions to drug pharmacokinetics and toxicity. Science 363, eaat9931 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Ackerman, D. & Schutze, H. The formation of trimethylamine by Bacterium prodigiosum. Zentralb. Physiol. 24, 210–211 (1910).

    Google Scholar 

  158. Romano, K. A., Vivas, E. I., Amador-Noguez, D. & Rey, F. E. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. Mbio 6, e02481 (2015).

    PubMed  PubMed Central  Google Scholar 

  159. Gregory, J. C. et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J. Biol. Chem. 290, 5647–5660 (2015).

    CAS  PubMed  Google Scholar 

  160. Guo, C.-J. et al. Discovery of reactive microbiota-derived metabolites that inhibit host proteases. Cell 168, 517–526.e18 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. McDonald, L. C. et al. Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin. Infect. Dis. 66, e1–e48 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Mullish, B. H. et al. The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut 67, 1920 (2018).

    PubMed  Google Scholar 

  164. DeFilipp, Z. et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. New Engl. J. Med. 381, 2043–2050 (2019).

    PubMed  Google Scholar 

  165. Korem, T. et al. Bread affects clinical parameters and induces gut microbiome-associated personal glycemic responses. Cell Metab. 25, 1243–1253.e5 (2017).

    CAS  PubMed  Google Scholar 

  166. Zeevi, D. et al. Personalized nutrition by prediction of glycemic responses. Cell 163, 1079–1094 (2015).

    CAS  PubMed  Google Scholar 

  167. Zhang, L. S. & Davies, S. S. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 8, 46 (2016).

    PubMed  PubMed Central  Google Scholar 

  168. Williams, B. B. et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 16, 495–503 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Saito, Y., Sato, T., Nomoto, K. & Tsuji, H. Identification of phenol- and p-cresol-producing intestinal bacteria by using media supplemented with tyrosine and its metabolites. Fems. Microbiol. Ecol. 94, fiy125 (2018).

    CAS  PubMed Central  Google Scholar 

  170. Eyssen, H., Pauw, G. D., Stragier, J. & Verhulst, A. Cooperative formation of ω-muricholic acid by intestinal microorganisms. Appl. Env. Microb. 45, 141–147 (1983).

    CAS  Google Scholar 

  171. Madsen, D., Beaver, M., Chang, L., Bruckner-Kardoss, E. & Wostmann, B. Analysis of bile acids in conventional and germfree rats. J. Lipid Res. 17, 107–111 (1976).

    CAS  PubMed  Google Scholar 

  172. Ragsdale, S. W. & Pierce, E. Acetogenesis and the Wood–Ljungdahl pathway of CO2 fixation. Biochim. Biophys. Acta 1784, 1873–1898 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Duncan, S. H., Barcenilla, A., Stewart, C. S., Pryde, S. E. & Flint, H. J. Acetate utilization and butyryl coenzyme A (CoA):acetate-CoA transferase in butyrate-producing bacteria from the human large intestine. Appl. Env. Microb. 68, 5186–5190 (2002).

    CAS  Google Scholar 

  174. Cloutier, N. et al. Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration. Proc. Natl Acad. Sci. USA 115, 201720553 (2018).

    Google Scholar 

  175. Fiehn, O. Metabolomics by gas chromatography–mass spectrometry: combined targeted and untargeted profiling. Curr. Protoc. Mol. Biol. 114, 30.4.1–30.4.32 (2016).

    Google Scholar 

  176. Berry, D. & Loy, A. Stable-isotope probing of human and animal microbiome function. Trends Microbiol. 26, 999–1007 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Röth, D. et al. Two-carbon folate cycle of commensal Lactobacillus reuteri 6475 gives rise to immunomodulatory ethionine, a source for histone ethylation. FASEB J. 33, 3536–3548 (2019).

    PubMed  Google Scholar 

  178. Jang, C. et al. The small intestine converts dietary fructose into glucose and organic acids. Cell Metab. 27, 351–361.e3 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Rath, C. M. et al. Molecular analysis of model gut microbiotas by imaging mass spectrometry and nanodesorption electrospray ionization reveals dietary metabolite transformations. Anal. Chem. 84, 9259–9267 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Wagner, M. Single-cell ecophysiology of microbes as revealed by Raman microspectroscopy or secondary ion mass spectrometry imaging. Annu. Rev. Microbiol. 63, 411–429 (2009).

    CAS  PubMed  Google Scholar 

  181. Lourenço, C. et al. Monitoring type 2 diabetes from volatile faecal metabolome in Cushing’s syndrome and single Afmid mouse models via a longitudinal study. Sci. Rep. 9, 18779 (2019).

    PubMed  PubMed Central  Google Scholar 

  182. Hoving, L. R., Heijink, M., Harmelen, V., Dijk, K. W. van & Giera, M. Methods in molecular biology. Meth. Mol. Biol. 1730, 247–256 (2018).

    CAS  Google Scholar 

  183. Zhang, X.-S. et al. Antibiotic-induced acceleration of type 1 diabetes alters maturation of innate intestinal immunity. eLife 7, e1002358 (2018).

    Google Scholar 

  184. Robinson, J. I. et al. Metabolomic networks connect host–microbiome processes to human Clostridioides difficile infections. J. Clin. Invest. 129, 3792–3806 (2019).

    PubMed  PubMed Central  Google Scholar 

  185. Fujisaka, S. et al. Diet, genetics, and the gut microbiome drive dynamic changes in plasma metabolites. Cell Rep. 22, 3072–3086 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Zierer, J. et al. The fecal metabolome as a functional readout of the gut microbiome. Nat. Genet. 50, 790–795 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Paul, H. A., Bomhof, M. R., Vogel, H. J. & Reimer, R. A. Diet-induced changes in maternal gut microbiota and metabolomic profiles influence programming of offspring obesity risk in rats. Sci. Rep. 6, 20683 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Lamichhane, S. et al. Strategy for nuclear-magnetic-resonance-based metabolomics of human feces. Anal. Chem. 87, 5930–5937 (2015).

    CAS  PubMed  Google Scholar 

  189. Zhang, C. et al. Dietary modulation of gut microbiota contributes to alleviation of both genetic and simple obesity in children. Ebiomedicine 2, 968–984 (2015).

    PubMed  PubMed Central  Google Scholar 

  190. Bui, T. P. N. et al. Production of butyrate from lysine and the Amadori product fructoselysine by a human gut commensal. Nat. Commun. 6, 10062 (2015).

    CAS  PubMed  Google Scholar 

  191. Lin, Y. et al. NMR-based fecal metabolomics fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget 7, 29454–29464 (2015).

    Google Scholar 

  192. Kim, M., Qie, Y., Park, J. & Kim, C. H. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe 20, 202–214 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Etchegaray, J.-P. & Mostoslavsky, R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes. Mol. Cell 62, 695–711 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Krautkramer, K. A., Rey, F. E. & Denu, J. M. Chemical signaling between gut microbiota and host chromatin: what is your gut really saying? J. Biol. Chem. 292, 8582–8593 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Allen, J. & Sears, C. L. Impact of the gut microbiome on the genome and epigenome of colon epithelial cells: contributions to colorectal cancer development. Genome Med. 11, 11 (2019).

    PubMed  PubMed Central  Google Scholar 

  196. Fan, J., Krautkramer, K. A., Feldman, J. L. & Denu, J. M. Metabolic regulation of histone post-translational modifications. ACS Chem. Biol. 10, 95–108 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Zhao, Y. & Garcia, B. A. Comprehensive catalog of currently documented histone modifications. Cold Spring Harb. Perspect. Biol. 7, a025064 (2015).

    PubMed  PubMed Central  Google Scholar 

  198. Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001).

    CAS  PubMed  Google Scholar 

  199. Badeaux, A. I. & Shi, Y. Emerging roles for chromatin as a signal integration and storage platform. Nat. Rev. Mol. Cell Bio. 14, 211 (2013).

    CAS  Google Scholar 

  200. Zhu, Q., Stöger, R. & Alberio, R. A lexicon of DNA modifications: their roles in embryo development and the germline. Front. Cell Dev. Biol. 6, 24 (2018).

    PubMed  PubMed Central  Google Scholar 

  201. Riggs, M. G., Whittaker, R. G., Neumann, J. R. & Ingram, V. M. n-Butyrate causes histone modification in HeLa and Friend erythroleukaemia cells. Nature 268, 462–464 (1977).

    CAS  PubMed  Google Scholar 

  202. Thaiss, C. A. et al. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 167, 1495–1510.e12 (2016).

    CAS  PubMed  Google Scholar 

  203. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Sánchez-Romero, M. A. & Casadesús, J. The bacterial epigenome. Nat. Rev. Microbiol. 18, 7–20 (2020).

    PubMed  Google Scholar 

  205. Dorman, C. J. & Dorman, M. J. DNA supercoiling is a fundamental regulatory principle in the control of bacterial gene expression. Biophys. Rev. 8, 89–100 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Liu, S. et al. The host shapes the gut microbiota via fecal microRNA. Cell Host Microbe 19, 32–43 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Yassour, M. et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 8, 343ra81–343ra81 (2016).

    PubMed  PubMed Central  Google Scholar 

  208. Kimura, I. et al. Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science 367, eaaw8429 (2020).

    CAS  PubMed  Google Scholar 

  209. Waterland, R. A. et al. Season of conception in rural gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet. 6, e1001252 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Romano, K. A. et al. Metabolic, epigenetic, and transgenerational effects of gut bacterial choline consumption. Cell Host Microbe 22, 279–290.e7 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

K.A.K acknowledges support from the Fulbright United States Scholar Program (US Fulbright Scholar Award) and the Human Frontier Science Program (HFSP; Long-term Fellowship LT000195/2018-L). Work in the Bäckhed laboratory is supported by Transatlantic Networks of Excellence Award from the Leducq Foundation (17CVD01), JPI (A healthy diet for a healthy life; 2017-01996_3), AFA insurances, the Swedish Research Council (2019-01599), the Swedish Heart Lung Foundation (20180600), the Knut and Alice Wallenberg Foundation (2017.0026), the Novo Nordisk Foundation (NNF19OC0057271, NNF17OC0028232 and NNF15OC0016798), grants from the Swedish state under the agreement between the Swedish government and the county councils, and the ALF-agreement (ALFGBG-718101). F.B. is Torsten Söderberg Professor in Medicine and recipient of an ERC Consolidator Grant (European Research Council; Consolidator grant 615362-METABASE).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed substantially to the conceptualization and writing of the manuscript, focusing on the following sections: Introduction (K.A.K and F.B.), Fermentable substrates (K.A.K. and F.B.), Amino acids and their derivatives (K.A.K. and F.B.), Bile acids as gut microbial messengers (K.A.K. and F.B), Vitamins and one-carbon metabolites (K.A.K. and F.B.), Perspectives (K.A.K., J.F., and F.B.), Box 1 (K.A.K., J.F., and F.B.) and Box 2 (K.A.K. and F.B.)

Corresponding author

Correspondence to Fredrik Bäckhed.

Ethics declarations

Competing interests

F.B. receives research support from Biogaia AB and is founder and shareholder in Implexion pharma AB. All other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Microbiology thanks P. Dorrestein, E. Gentry 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.

Glossary

Mucosa

An epithelial layer comprising epithelial cells and mucus-secreting cells, among other specialized cell types, that lines multiple body surfaces (gastrointestinal tract, oropharynx, airways and vaginal tract) and functions as an innate barrier.

Fermentation

The chemical breakdown of organic substrates (for example, carbohydrates and amino acids) by various enzymes in the absence of molecular oxygen.

Gas chromatography–mass spectrometry

(GC-MS). An analytical method that couples chromatographic separation of complex biological samples in the gas phase to mass spectrometry for the identification and quantification of the compounds that comprise the sample.

Liquid chromatography–mass spectrometry

(LC-MS). An analytical method that couples chromatographic separation of complex biological samples in the liquid phase to mass spectrometry for the identification and quantification of the compounds that comprise the sample.

Desorption electrospray ionization mass spectrometry

A soft electrospray ionization technique that relies on solvent extraction directly on the sample under ambient conditions that is primarily used on tissues for imaging mass spectrometry.

Raman spectroscopy

A vibrational spectroscopy technique wherein a biological sample is subjected to a beam of light and differences in photon scatter (based on the molecular composition of the sample) are used to produce a unique chemical fingerprint.

Carbohydrate-active enzymes

(CAZymes). A collective term for enzymes that can synthesize or break down saccharides.

Microbial accessible carbohydrates

(MACs). Complex polysaccharides and oligosaccharides that are available to the gut microbiome’s vast repertoire of carbohydrate-active enzymes.

Mucus

A gel-like layer(s) secreted by and resting on top of the mucosa comprising mucins and functions as an essential barrier between the environment and the mucosal layer.

Mucins

Large, heavily decorated proteins characterized by proline-rich, serine-rich and threonine-rich tandem repeats (PTS domains) that are modified by complex O-glycans and form large polymeric protein networks that function as the building blocks of mucus in the intestinal tract.

Exoglycosidases

Enzymes that hydrolyse the glycosidic bond at the terminal monosaccharide in a polysaccharide or oligosaccharide.

Polysaccharidases

Enzymes that hydrolyse polysaccharides to form smaller saccharide chains.

Glycosidases

A general term for enzymes that hydrolyse glycosidic bonds in polysaccharides and oligosaccharides.

Chromatin

A highly structured nucleoprotein complex in eukaryotes that consists of the nucleic acids and histone proteins around which double-stranded genomic DNA winds to ultimately form chromosomes.

Germ-free mice

Mice born and raised in the complete absence of any microorganisms, frequently in a laminar flow glovebox isolator or IsoCage setting

Conventionally raised mice

Mice born and raised in a normal (‘conventional’) mouse colony setting with exposure to normal environmental microorganisms from birth onwards.

Bile salt hydrolases

Microbial enzymes that hydrolyse the amide bond in taurine and glycine-conjugated primary bile acids to yield a deconjugated bile acid.

Nuclear magnetic resonance spectroscopy

An analytical method frequently used in structural, quantitative and imaging applications wherein unique spectra are obtained for biomolecules based on nuclear resonance transitions that occur when atomic nuclei are immersed in a magnetic field and then subjected to specific magnetic energy levels.

Faecal microbiota transplantation

Delivery of processed stool from a donor into the intestinal tract of a recipient with the goal of stable engraftment.

Enterotypes

Variants in human microbial community composition based on empirical population measurements that are dominated by a single genus (for example, Bacteroides, Ruminococcus or Prevotella).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Krautkramer, K.A., Fan, J. & Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol 19, 77–94 (2021). https://doi.org/10.1038/s41579-020-0438-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-020-0438-4

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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