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:

Short-chain fatty acids in control of body weight and insulin sensitivity

Key Points

  • Short-chain fatty acids (SCFA), which are derived from gut microbial fermentation of indigestible foods, have important metabolic functions and are crucial for intestinal health

  • The discovery of SCFA receptors in many different tissues highlights that SCFA are involved in the crosstalk between the gut and peripheral tissues

  • In addition to their role in gut health and as signalling molecules, SCFA might enter the systemic circulation and directly affect substrate metabolism and function of peripheral tissues

  • SCFA might increase intestinal energy harvesting and promote the development of obesity, but could also increase energy expenditure and anorexic hormone production, as well as improving appetite regulation

  • Increasing evidence supports a beneficial role for SCFA in adipose tissue, skeletal muscle and liver substrate metabolism and function, thereby contributing to improved insulin sensitivity

  • Well-controlled human intervention studies investigating the role of SCFA and differential SCFA availability on gut and systemic metabolic health are eagerly awaited

Abstract

The connection between the gut microbiota and the aetiology of obesity and cardiometabolic disorders is increasingly being recognized by clinicians. Our gut microbiota might affect the cardiometabolic phenotype by fermenting indigestible dietary components and thereby producing short-chain fatty acids (SCFA). These SCFA are not only of importance in gut health and as signalling molecules, but might also enter the systemic circulation and directly affect metabolism or the function of peripheral tissues. In this Review, we discuss the effects of three SCFA (acetate, propionate and butyrate) on energy homeostasis and metabolism, as well as how these SCFA can beneficially modulate adipose tissue, skeletal muscle and liver tissue function. As a result, these SCFA contribute to improved glucose homeostasis and insulin sensitivity. Furthermore, we also summarize the increasing evidence for a potential role of SCFA as metabolic targets to prevent and counteract obesity and its associated disorders in glucose metabolism and insulin resistance. However, most data are derived from animal and in vitro studies, and consequently the importance of SCFA and differential SCFA availability in human energy and substrate metabolism remains to be fully established. Well-controlled human intervention studies investigating the role of SCFA on cardiometabolic health are, therefore, eagerly awaited.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Interorgan crosstalk and obesity-induced insulin resistance.
Figure 2: SCFA and interorgan crosstalk.
Figure 3: The effect of SCFA on adipose tissue function.
Figure 4: SCFA and skeletal muscle substrate metabolism.
Figure 5: SCFA and liver function.

Similar content being viewed by others

References

  1. Shulman, G. I. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N. Engl. J. Med. 371, 1131–1141 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Grundy, S. M. Obesity, metabolic syndrome, and cardiovascular disease. J. Clin. Endocrinol. Metab. 89, 2595–2600 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Bäckhed, F. et al. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl Acad. Sci. USA 101, 15718–15723 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Membrez, M. et al. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J. 22, 2416–2426 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Andersson, U. et al. Probiotics lower plasma glucose in the high-fat fed C57BL/6J mouse. Benef. Microbes 1, 189–196 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Vrieze, A. et al. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J. Hepatol. 60, 824–831 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  10. Topping, D. L. & Clifton, P. M. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 81, 1031–1064 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Gao, Z. et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58, 1509–1517 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lin, H. V. et al. Butyrate and propionate protect. against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS ONE 7, e35240 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Frost, G. et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 5, 3611 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Freeland, K. R. & Wolever, T. Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-alpha. Br. J. Nutr. 103, 460–466 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. den Besten, G. et al. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes 64, 2398–2408 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Murphy, E. et al. Composition and energy harvesting capacity of the gut microbiota: relationship to diet, obesity and time in mouse models. Gut 59, 1635–1642 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Al-Lahham, S. H. et al. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur. J. Clin. Invest. 42, 357–364 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Al-Lahham, S. H. et al. Regulation of adipokine production in human adipose tissue by propionic acid. Eur. J. Clin. Invest. 40, 401–407 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Macfarlane, G. T. & Macfarlane, S. Bacteria, colonic fermentation, and gastrointestinal health. J. AOAC Int. 95, 50–60 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Blachier, F., Mariotti, F., Huneau, J. & Tome, D. Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids 33, 547–562 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Geypens, B. et al. Influence of dietary protein supplements on the formation of bacterial metabolites in the colon. Gut 41, 70–76 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Macfarlane, G., Gibson, G., Beatty, E. & Cummings, J. 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).

    Article  CAS  Google Scholar 

  25. Corpet, D. E. et al. Colonic protein fermentation and promotion of colon carcinogenesis by thermolyzed casein. Nutr. Cancer 23, 271–281 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Windey, K., De Preter, V. & Verbeke, K. Relevance of protein fermentation to gut health. Mol. Nutr. Food Res. 56, 184–196 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Wong, J. M., de Souza, R., Kendall, C. W., Emam, A. & Jenkins, D. J. Colonic health: fermentation and short chain fatty acids. J. Clin. Gastroenterol. 40, 235–243 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Schwiertz, A. et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 18, 190–195 (2009).

    Article  PubMed  Google Scholar 

  29. Fernandes, J., Su, W., Rahat-Rozenbloom, S., Wolever, T. & Comelli, E. Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr. Diabetes 4, e121 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cummings, J. H. & Macfarlane, G. T. Colonic microflora: nutrition and health. Nutrition 13, 476–478 (1997).

    Article  CAS  PubMed  Google Scholar 

  31. McBurney, M. I. & Thompson, L. U. In vitro fermentabilities of purified fiber supplements. J. Food Sci. 54, 347–350 (1989).

    Article  Google Scholar 

  32. McBurney, M., Thompson, L., Cuff, D. & Jenkins, D. Comparison of ileal effluents, dietary fibers, and whole foods in predicting the physiological importance of colonic fermentation. Am. J. Gastroenterol. 83, 536–540 (1988).

    CAS  PubMed  Google Scholar 

  33. Venema, K. Microbial metabolites produced by the colonic microbiota as drivers for immunomodulation in the host. FASEB J. 27, 643.12 (2013).

    Google Scholar 

  34. Pylkas, A., Juneja, L. & Slavin, J. Comparison of different fibers for in vitro production of short chain fatty acids by intestinal microflora. J. Med. Food 8, 113–116 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Lampe, J., Fredstrom, S., Slavin, J. & Potter, J. Sex differences in colonic function: a randomised trial. Gut 34, 531–536 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lampe, J., Wetsch, R., Thompson, W. & Slavin, J. Gastrointestinal effects of sugarbeet fiber and wheat bran in healthy men. Eur. J. Clin. Nutr. 47, 543–548 (1993).

    CAS  PubMed  Google Scholar 

  37. Owens, F. & Isaacson, H. Ruminal microbial yields: factors influencing synthesis and bypass. Fed. Proc. 36, 198–202 (1977).

    CAS  PubMed  Google Scholar 

  38. Lewis, S. & Heaton, K. Increasing butyrate concentration in the distal colon by accelerating intestinal transit. Gut 41, 245–251 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. El Oufir, L. et al. Relationships between transit time in man and in vitro fermentation of dietary fiber by fecal bacteria. Eur. J. Clin. Nutr. 54, 603–609 (2000).

    Article  Google Scholar 

  40. Ruppin, H., Bar-Meir, S., Soergel, K., Wood, C. & Schmitt, M. Jr. Absorption of short-chain fatty acids by the colon. Gastroenterology 78, 1500–1507 (1980).

    Article  CAS  PubMed  Google Scholar 

  41. Titus, E. & Ahearn, G. A. Short-chain fatty acid transport in the intestine of a herbivorous teleost. J. Exp. Biol. 135, 77–94 (1988).

    CAS  PubMed  Google Scholar 

  42. Ritzhaupt, A., Wood, I. S., Ellis, A., Hosie, K. B. & Shirazi-Beechey, S. P. Identification and characterization of a monocarboxylate transporter (MCT1) in pig and human colon: its potential to transport L-lactate as well as butyrate. J. Physiol. 513, 719–732 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Moschen, I., Bröer, A., Galic, S., Lang, F. & Bröer, S. Significance of short chain fatty acid transport by members of the monocarboxylate transporter family (MCT). Neurochem. Res. 37, 2562–2568 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Bloemen, J. G. et al. Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin. Nutr. 28, 657–661 (2009).

    Article  CAS  PubMed  Google Scholar 

  45. Araghizadeh, A. & Abdelnaby, A. in Colorectal Surgery (eds Bailey, H. R., Billingham, R. P., Stamos, M. J. & Snyder, M. J.) 3–17 (Elsevier Health Sciences, 2012).

    Google Scholar 

  46. Bloemen, J. G. et al. Short chain fatty acids exchange: Is the cirrhotic, dysfunctional liver still able to clear them? Clin. Nutr. 29, 365–369 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Robertson, M. D., Bickerton, A. S., Dennis, A. L., Vidal, H. & Frayn, K. N. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am. J. Clin. Nutr. 82, 559–567 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Roediger, W. Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 21, 793–798 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Roediger, W. E. W. in Physiological and Clinical Aspects of Short-Chain Fatty Acids (eds Cummings, J. H., Rombeau, J. L. & Sakata, T.) 337–351 (Cambridge University Press, 1995).

    Google Scholar 

  50. Ardawi, M. & Newsholme, E. Fuel utilization in colonocytes of the rat. Biochem. J. 231, 713–719 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Frankel, W. et al. Mediation of the trophic effects of short-chain fatty acids on the rat jejunum and colon. Gastroenterology 106, 375–380 (1994).

    Article  CAS  PubMed  Google Scholar 

  52. Brown, A. J. et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 278, 11312–11319 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Le Poul, E. et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 278, 25481–25489 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Karaki, S. et al. Expression of the short-chain fatty acid receptor, GPR43, in the human colon. J. Mol. Histol. 39, 135–142 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Tazoe, H. et al. Expression of short-chain fatty acid receptor GPR41 in the human colon. Biomed. Res. 30, 149–156 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Thangaraju, M. et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 69, 2826–2832 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Liu, F. et al. The expression of GPR109A, NF-κB and IL-1β in peripheral blood leukocytes from patients with type 2 diabetes. Ann. Clin. Lab. Sci. 44, 443–448 (2014).

    CAS  PubMed  Google Scholar 

  58. Taggart, A. K. et al. (D)-β-hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J. Biol. Chem. 280, 26649–26652 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Tunaru, S. et al. PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat. Med. 9, 352–355 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Pluznick, J. L. et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl Acad. Sci. USA 110, 4410–4415 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Hellman, B., Larsson, S. & Westman, S. Acetate metabolism in isolated epididymal adipose tissue from obese-hyperglycemic mice of different ages. Acta Physiol. Scand. 56, 189–198 (1962).

    Article  CAS  PubMed  Google Scholar 

  62. Villee, C. The effect of insulin on the incorporation of C14-labeled pyruvate and acetate into lipid and protein. Anat. Rec. 101, 680 (1948).

    CAS  PubMed  Google Scholar 

  63. Feller, D. Metabolism of adipose tissue. I. Incorporation of acetate carbon into lipides by slices of adipose tissue. J. Biol. Chem. 206, 171–180 (1954).

    CAS  PubMed  Google Scholar 

  64. Elwood, J., Marcó, A. & Van Bruggen, J. Lipid metabolism in the diabetic rat. IV. Metabolism of acetate, acetoacetate, butyrate, and mevalonate in vitro. J. Biol. Chem. 235, 573–577 (1960).

    CAS  PubMed  Google Scholar 

  65. Wong, R. K. & Van Bruggen, J. Lipid metabolism in the diabetic rat. I. Acetate metabolism and lipid synthesis in vivo. J. Biol. Chem. 235, 26–29 (1960).

    CAS  PubMed  Google Scholar 

  66. Reshef, L., Niv, J. & Shapiro, B. Effect of propionate on lipogenesis in adipose tissue. J. Lipid Res. 8, 682–687 (1967).

    CAS  PubMed  Google Scholar 

  67. Sakata, T. Effects of indigestible dietary bulk and short chain fatty acids on the tissue weight and epithelial cell proliferation rate of the digestive tract in rats. J. Nutr. Sci. Vitaminol. (Tokyo) 32, 355–362 (1986).

    Article  CAS  Google Scholar 

  68. Boillot, J. et al. Effects of dietary propionate on hepatic glucose production, whole-body glucose utilization, carbohydrate and lipid metabolism in normal rats. Br. J. Nutr. 73, 241–251 (1995).

    Article  CAS  PubMed  Google Scholar 

  69. Crouse, J. R., Gerson, C. D., DeCarli, L. M. & Lieber, C. S. Role of acetate in the reduction of plasma free fatty acids produced by ethanol in man. J. Lipid Res. 9, 509–512 (1968).

    CAS  PubMed  Google Scholar 

  70. Björntorp, P. & Hood, B. Studies on adipose tissue from obese patients with or without diabetes mellitus. Acta Med. Scand. 179, 221–227 (1966).

    Article  PubMed  Google Scholar 

  71. Todesco, T., Rao, A. V., Bosello, O. & Jenkins, D. Propionate lowers blood glucose and alters lipid metabolism in healthy subjects. Am. J. Clin. Nutr. 54, 860–865 (1991).

    Article  CAS  PubMed  Google Scholar 

  72. Wolever, T., Brighenti, F., Royall, D., Jenkins, A. & Jenkins, D. Effect of rectal infusion of short chain fatty acids in human subjects. Am. J. Gastroenterol. 84, 1027–1033 (1989).

    CAS  PubMed  Google Scholar 

  73. Zoetendal, E. G., Collier, C. T., Koike, S., Mackie, R. I. & Gaskins, H. R. Molecular ecological analysis of the gastrointestinal microbiota: a review. J. Nutr. 134, 465–472 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Muyzer, G., De Waal, E. C. & Uitterlinden, A. G. Profiling of complex microbial populations by denaturing gradient gel electrophoresis analysis of polymerase chain reaction-amplified genes coding for 16S rRNA. Appl. Environ. Microbiol. 59, 695–700 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhao, L. The gut microbiota and obesity: from correlation to causality. Nat. Rev. Microbiol. 11, 639–647 (2013).

    Article  CAS  PubMed  Google Scholar 

  76. Hartstra, A. V., Bouter, K. E., Bäckhed, F. & Nieuwdorp, M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care 38, 159–165 (2015).

    Article  CAS  PubMed  Google Scholar 

  77. Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pedersen, M. H., Lauritzen, L. & Hellgren, L. I. Fish oil combined with SCFA synergistically prevent tissue accumulation of NEFA during weight loss in obese mice. Br. J. Nutr. 106, 1449–1456 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  80. Fujii, H. et al. Impact of dietary fiber intake on glycemic control, cardiovascular risk factors and chronic kidney disease in Japanese patients with type 2 diabetes mellitus: the Fukuoka Diabetes Registry. Nutr. J. 12, 159 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Konings, E., Schoffelen, P. F., Stegen, J. & Blaak, E. E. Effect of polydextrose and soluble maize fibre on energy metabolism, metabolic profile and appetite control in overweight men and women. Br. J. Nutr. 111, 111–121 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Reichert, R. G. et al. Decreasing cardiovascular risk factors in obese individuals using a combination of PGX® meal replacements and PGX® granules in a 12-week clinical weight modification program. J. Complement. Integr. Med. 10, 135–142 (2013).

    Article  Google Scholar 

  83. Hashizume, C. et al. Improvement effect of resistant maltodextrin in humans with metabolic syndrome by continuous administration. J. Nutr. Sci. Vitaminol. 58, 423–430 (2012).

    Article  CAS  PubMed  Google Scholar 

  84. Cani, P. D. et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am. J. Clin. Nutr. 90, 1236–1243 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Cani, P. D., Joly, E., Horsmans, Y. & Delzenne, N. M. Oligofructose promotes satiety in healthy human: a pilot study. Eur. J. Clin. Nutr. 60, 567–572 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Vulevic, J., Juric, A., Tzortzis, G. & Gibson, G. R. A mixture of trans-galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal microbiota and immune function of overweight adults. J. Nutr. 143, 324–331 (2013).

    Article  CAS  PubMed  Google Scholar 

  87. Daud, N. M. et al. The impact of oligofructose on stimulation of gut hormones, appetite regulation and adiposity. Obesity 22, 1430–1438 (2014).

    Article  CAS  PubMed  Google Scholar 

  88. Dewulf, E. et al. Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 62, 1112–1121 (2013).

    Article  CAS  PubMed  Google Scholar 

  89. Parnell, J. A. & Reimer, R. A. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am. J. Clin. Nutr. 89, 1751–1759 (2009).

    Article  CAS  PubMed  Google Scholar 

  90. Royall, D., Wolever, T. & Jeejeebhoy, K. N. Clinical significance of colonic fermentation. Am. J. Gastroenterol. 85, 1307–1312 (1990).

    CAS  PubMed  Google Scholar 

  91. Alhabeeb, H., Chambers, E., Frost, G., Morrison, D. & Preston, T. Inulin propionate ester increases satiety and decreases appetite but does not affect gastric emptying in healthy humans. Proc. Nutr. Soc. 73, E21 (2014).

    Article  Google Scholar 

  92. Chambers, E. S. et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut http://dx.doi.org/10.1136/gutjnl-2014-307913.

  93. Kondo, T., Kishi, M., Fushimi, T., Ugajin, S. & Kaga, T. Vinegar intake reduces body weight, body fat mass, and serum triglyceride levels in obese Japanese subjects. Biosci. Biotechnol. Biochem. 73, 1837–1843 (2009).

    Article  CAS  PubMed  Google Scholar 

  94. Theodorakis, M. J. et al. Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1 and GIP. Am. J. Physiol. Endocrinol. Metab. 290, E550–E559 (2006).

    Article  CAS  PubMed  Google Scholar 

  95. Roberge, J. N. & Brubaker, P. L. Regulation of intestinal proglucagon-derived peptide secretion by glucose-dependent insulinotropic peptide in a novel enteroendocrine loop. Endocrinology 133, 233–240 (1993).

    Article  CAS  PubMed  Google Scholar 

  96. De Silva, A. & Bloom, S. R. Gut hormones and appetite control: a focus on PYY and GLP-1 as therapeutic targets in obesity. Gut Liver 6, 10–20 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Savage, A., Adrian, T., Carolan, G., Chatterjee, V. & Bloom, S. Effects of peptide YY (PYY) on mouth to caecum intestinal transit time and on the rate of gastric emptying in healthy volunteers. Gut 28, 166–170 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Murphy, K. G. & Bloom, S. R. Gut hormones and the regulation of energy homeostasis. Nature 444, 854–859 (2006).

    Article  CAS  PubMed  Google Scholar 

  99. Näslund, E. et al. GLP-1 slows solid gastric emptying and inhibits insulin, glucagon, and PYY release in humans. Am. J. Physiol. 277, R910–R916 (1999).

    PubMed  Google Scholar 

  100. Schjoldager, B., Mortensen, P., Christiansen, J., Orskov, C. & Holst, J. GLP-1 (glucagon-like peptide 1) and truncated GLP-1, fragments of human proglucagon, inhibit gastric acid secretion in humans. Dig. Dis. Sci. 34, 703–708 (1989).

    Article  CAS  PubMed  Google Scholar 

  101. Tolhurst, G. et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364–371 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Psichas, A. et al. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 39, 424–429 (2014).

    Article  CAS  Google Scholar 

  103. Reimer, R. A. et al. A human cellular model for studying the regulation of glucagon-like peptide-1 secretion. Endocrinology 142, 4522–4528 (2001).

    Article  CAS  PubMed  Google Scholar 

  104. Psichas, A. et al. Short chain fatty acids stimulate the release of gut hormone peptide YY from human primary enteroendocrine L cells. Proc. Physiol. Soc. 27, PC331 (2012).

    Google Scholar 

  105. Karaki, S. et al. Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 324, 353–360 (2006).

    Article  CAS  PubMed  Google Scholar 

  106. Cani, P. D., Dewever, C. & Delzenne, N. M. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br. J. Nutr. 92, 521–526 (2004).

    Article  CAS  PubMed  Google Scholar 

  107. Zhou, J. et al. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents. Am. J. Physiol. Endocrinol. Metab. 295, E1160–E1166 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Cani, P. D., Neyrinck, A. M., Maton, N. & Delzenne, N. M. Oligofructose promotes satiety in rats fed a high-fat diet: involvement of glucagon-like peptide-1. Obesity Res. 13, 1000–1007 (2005).

    Article  CAS  Google Scholar 

  109. Zhou, J. et al. Peptide YY and proglucagon mRNA expression patterns and regulation in the gut. Obesity 14, 683–689 (2006).

    Article  CAS  PubMed  Google Scholar 

  110. Cani, P. D., Hoste, S., Guiot, Y. & Delzenne, N. M. Dietary non-digestible carbohydrates promote L-cell differentiation in the proximal colon of rats. Br. J. Nutr. 98, 32–37 (2007).

    Article  CAS  PubMed  Google Scholar 

  111. Pedersen, C. et al. Gut hormone release and appetite regulation in healthy non-obese participants following oligofructose intake. A dose-escalation study. Appetite 66, 44–53 (2013).

    Article  PubMed  Google Scholar 

  112. Wichmann, A. et al. Microbial modulation of energy availability in the colon regulates intestinal transit. Cell Host Microbe 14, 582–590 (2013).

    Article  CAS  PubMed  Google Scholar 

  113. Jiang, L. et al. Increased brain uptake and oxidation of acetate in heavy drinkers. J. Clin. Invest. 123, 1605–1614 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Xiong, Y. et al. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl Acad. Sci. USA 101, 1045–1050 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Soliman, M. et al. Inverse regulation of leptin mRNA expression by short-and long-chain fatty acids in cultured bovine adipocytes. Domest. Anim. Endocrinol. 33, 400–409 (2007).

    Article  CAS  PubMed  Google Scholar 

  116. Lee, S. & Hossner, K. Coordinate regulation of ovine adipose tissue gene expression by propionate. J. Anim. Sci. 80, 2840–2849 (2002).

    Article  CAS  PubMed  Google Scholar 

  117. Ioffe, E., Moon, B., Connolly, E. & Friedman, J. M. Abnormal regulation of the leptin gene in the pathogenesis of obesity. Proc. Natl Acad. Sci. USA 95, 11852–11857 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Maffei, M. et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat. Med. 1, 1155–1161 (1995).

    Article  CAS  PubMed  Google Scholar 

  119. Frederich, R. C. et al. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat. Med. 1, 1311–1314 (1995).

    Article  CAS  PubMed  Google Scholar 

  120. Kondo, T., Kishi, M., Fushimi, T. & Kaga, T. Acetic acid upregulates the expression of genes for fatty acid oxidation enzymes in liver to suppress body fat accumulation. J. Agric. Food Chem. 57, 5982–5986 (2009).

    Article  CAS  PubMed  Google Scholar 

  121. Kimura, I. et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl Acad. Sci. USA 108, 8030–8035 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Mithieux, G. & Gautier-Stein, A. Intestinal glucose metabolism revisited. Diabetes Res. Clin. Pract. 105, 295–301 (2014).

    Article  CAS  PubMed  Google Scholar 

  123. Ge, H. et al. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology 149, 4519–4526 (2008).

    Article  CAS  PubMed  Google Scholar 

  124. Aberdein, N., Schweizer, M. & Ball, D. Sodium acetate decreases phosphorylation of hormone sensitive lipase in isoproterenol-stimulated 3T3-L1 mature adipocytes. Adipocyte 3, 121–125 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Rumberger, J. M., Arch, J. R. & Green, A. Butyrate and other short-chain fatty acids increase the rate of lipolysis in 3T3-L1 adipocytes. PeerJ 2, e611 (2013).

    Article  CAS  Google Scholar 

  126. Hong, Y. H. et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology 146, 5092–5099 (2005).

    Article  CAS  PubMed  Google Scholar 

  127. Kimura, I. et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 4, 1829 (2013).

    Article  CAS  PubMed  Google Scholar 

  128. Inoue, D., Tsujimoto, G. & Kimura, I. Regulation of energy homeostasis by GPR41. Front. Endocrinol. (Laussane) 5, 81 (2014).

    Google Scholar 

  129. Zaibi, M. S. et al. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett. 584, 2381–2386 (2010).

    Article  CAS  PubMed  Google Scholar 

  130. Frost, G. et al. Effect of short chain fatty acids on the expression of free fatty acid receptor 2 (Ffar2), Ffar3 and early-stage adipogenesis. Nutr. Diabetes 4, e128 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Merkel, M., Eckel, R. H. & Goldberg, I. J. Lipoprotein lipase genetics, lipid uptake, and regulation. J. Lipid Res. 43, 1997–2006 (2002).

    Article  CAS  PubMed  Google Scholar 

  132. Grootaert, C. et al. Bacterial monocultures, propionate, butyrate and H2O2 modulate the expression, secretion and structure of the fasting induced adipose factor in gut epithelial cell lines. Environ. Microbiol. 13, 1778–1789 (2011).

    Article  CAS  PubMed  Google Scholar 

  133. Haberland, M., Carrer, M., Mokalled, M. H., Montgomery, R. L. & Olson, E. N. Redundant control of adipogenesis by histone deacetylases 1 and 2. J. Biol. Chem. 285, 14663–14670 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Li, G., Yao, W. & Jiang, H. Short-chain fatty acids enhance adipocyte differentiation in the stromal vascular fraction of porcine adipose tissue. J. Nutr. 144, 1887–1895 (2014).

    Article  CAS  PubMed  Google Scholar 

  135. Dewulf, E. M. et al. Inulin-type fructans with prebiotic properties counteract GPR43 overexpression and PPARγ-related adipogenesis in the white adipose tissue of high-fat diet-fed mice. J. Nutr. Biochem. 22, 712–722 (2011).

    Article  CAS  PubMed  Google Scholar 

  136. Dewulf, E. M. et al. Evaluation of the relationship between GPR43 and adiposity in human. Nutr. Metab. 10, 11 (2013).

    Article  CAS  Google Scholar 

  137. Goossens, G. H. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. Physiol. Behav. 94, 206–218 (2008).

    Article  CAS  PubMed  Google Scholar 

  138. Tilg, H. & Moschen, A. R. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat. Rev. Immunol. 6, 772–783 (2006).

    Article  CAS  PubMed  Google Scholar 

  139. Apovian, C. M. et al. Adipose macrophage infiltration is associated with insulin resistance and vascular endothelial dysfunction in obese subjects. Arterioscler. Thromb. Vasc. Biol. 28, 1654–1659 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Chaudhry, A. & Rudensky, A. Y. Control of inflammation by integration of environmental cues by regulatory T cells. J. Clin. Invest. 123, 939–944 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  142. Mazurek, T. et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation 108, 2460–2466 (2003).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  144. Furusawa, Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).

    Article  CAS  PubMed  Google Scholar 

  145. Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Mariadason, J. M., Barkla, D. H. & Gibson, P. R. Effect of short-chain fatty acids on paracellular permeability in Caco-2 intestinal epithelium model. Am. J. Physiol. 272, G705–G712 (1997).

    CAS  PubMed  Google Scholar 

  147. Malago, J. et al. Differential modulation of enterocyte-like Caco-2 cells after exposure to short-chain fatty acids. Food Addit. Contam. 20, 427–437 (2003).

    Article  CAS  PubMed  Google Scholar 

  148. Suzuki, T., Yoshida, S. & Hara, H. Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability. Br. J. Nutr. 100, 297–305 (2008).

    Article  CAS  PubMed  Google Scholar 

  149. Elamin, E. E., Masclee, A. A., Dekker, J., Pieters, H.-J. & Jonkers, D. M. Short-chain fatty acids activate AMP-activated protein kinase and ameliorate ethanol-induced intestinal barrier dysfunction in Caco-2 cell monolayers. J. Nutr. 143, 1872–1881 (2013).

    Article  CAS  PubMed  Google Scholar 

  150. Macia, L. et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 6, 6734 (2015).

    Article  CAS  PubMed  Google Scholar 

  151. Cani, P. D. et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 50, 2374–2383 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  153. Mehta, N. N. et al. Experimental endotoxemia induces adipose inflammation and insulin resistance in humans. Diabetes 59, 172–181 (2010).

    Article  CAS  PubMed  Google Scholar 

  154. Vila, I. K. et al. Immune cell toll-like receptor 4 mediates the development of obesity-and endotoxemia-associated adipose tissue fibrosis. Cell Rep. 7, 1116–1129 (2014).

    Article  CAS  PubMed  Google Scholar 

  155. Muccioli, G. G. et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol. 6, 392 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Tedelind, S., Westberg, F., Kjerrulf, M. & Vidal, A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J. Gastroenterol. 13, 2826–2832 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Liu, T. et al. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation 35, 1676–1684 (2012).

    Article  CAS  PubMed  Google Scholar 

  158. Cox, M. A. et al. Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E2 and cytokines. World J. Gastroenterol. 15, 5549–5557 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Ohira, H. et al. Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J. Atheroscler. Thromb. 20, 425–442 (2013).

    Article  CAS  PubMed  Google Scholar 

  160. Koves, T. R. et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 7, 45–56 (2008).

    Article  CAS  PubMed  Google Scholar 

  161. Corpeleijn, E., Saris, W. & Blaak, E. Metabolic flexibility in the development of insulin resistance and type 2 diabetes: effects of lifestyle. Obes. Rev. 10, 178–193 (2009).

    Article  CAS  PubMed  Google Scholar 

  162. Yamashita, H. et al. Effects of acetate on lipid metabolism in muscles and adipose tissues of type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 73, 570–576 (2009).

    Article  CAS  PubMed  Google Scholar 

  163. Fushimi, T. et al. Acetic acid feeding enhances glycogen repletion in liver and skeletal muscle of rats. J. Nutr. 131, 1973–1977 (2001).

    Article  CAS  PubMed  Google Scholar 

  164. Bonini, J. A., Anderson, S. M. & Steiner, D. F. Molecular cloning and tissue expression of a novel orphan G protein-coupled receptor from rat lung. Biochem. Biophys. Res. Comm. 234, 190–193 (1997).

    Article  CAS  PubMed  Google Scholar 

  165. Chai, W. et al. Glucagon-like peptide 1 recruits microvasculature and increases glucose use in muscle via a nitric oxide–dependent mechanism. Diabetes 61, 888–896 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  167. Tazoe, H. et al. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions. J. Physiol. Pharmacol. 59, 251–262 (2008).

    PubMed  Google Scholar 

  168. 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).

    Article  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  170. Demigne, C. et al. Effect of propionate on fatty acid and cholesterol synthesis and on acetate metabolism in isolated rat hepatocytes. Br. J. Nutr. 74, 209–219 (1995).

    Article  CAS  PubMed  Google Scholar 

  171. Hardie, D. & Pan, D. Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem. Soc. Trans. 30, 1064–1070 (2002).

    Article  CAS  PubMed  Google Scholar 

  172. Zhang, B. B., Zhou, G. & Li, C. AMPK: an emerging drug target for diabetes and the metabolic syndrome. Cell Metab. 9, 407–416 (2009).

    Article  CAS  PubMed  Google Scholar 

  173. Li, X. et al. Acetic acid activates the AMP-activated protein kinase signaling pathway to regulate lipid metabolism in bovine hepatocytes. PLoS ONE 8, e67880 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Yamashita, H. et al. Improvement of obesity and glucose tolerance by acetate in type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 71, 1236–1243 (2007).

    Article  CAS  PubMed  Google Scholar 

  175. Sakakibara, S., Yamauchi, T., Oshima, Y., Tsukamoto, Y. & Kadowaki, T. Acetic acid activates hepatic AMPK and reduces hyperglycemia in diabetic KK-A(y) mice. Biochem. Biophys. Res. Comm. 344, 597–604 (2006).

    Article  CAS  PubMed  Google Scholar 

  176. Endo, H., Niioka, M., Kobayashi, N., Tanaka, M. & Watanabe, T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS ONE 8, e63388 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Wolever, T., Spadafora, P. & Eshuis, H. Interaction between colonic acetate and propionate in humans. Am. J. Clin. Nutr. 53, 681–687 (1991).

    Article  CAS  PubMed  Google Scholar 

  178. Laurent, C. et al. Effect of acetate and propionate on fasting hepatic glucose production in humans. Eur. J. Clin. Nutr. 49, 484–491 (1995).

    CAS  PubMed  Google Scholar 

  179. Fernandes, J., Vogt, J. & Wolever, T. M. Intravenous acetate elicits a greater free fatty acid rebound in normal than hyperinsulinaemic humans. Eur. J. Clin. Nutr. 66, 1029–1034 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Venter, C. S., Vorster, H. H. & Cummings, J. H. Effects of dietary propionate on carbohydrate and lipid metabolism in healthy volunteers. Am. J. Gastroenterol. 85, 549–553 (1990).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Research in the authors' laboratory is funded by TI Food and Nutrition, a public–private partnership on precompetitive research in food and nutrition.

Author information

Authors and Affiliations

Authors

Contributions

All authors researched the data for the article, discussed the content, wrote, edited and approved the manuscript before submission.

Corresponding author

Correspondence to Ellen E. Blaak.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Canfora, E., Jocken, J. & Blaak, E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 11, 577–591 (2015). https://doi.org/10.1038/nrendo.2015.128

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrendo.2015.128

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