The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents

A Psichas, M L Sleeth, K G Murphy, L Brooks, G A Bewick, A C Hanyaloglu, M A Ghatei, S R Bloom, G Frost, A Psichas, M L Sleeth, K G Murphy, L Brooks, G A Bewick, A C Hanyaloglu, M A Ghatei, S R Bloom, G Frost

Abstract

Background and objectives: The gut hormones peptide YY (PYY) and glucagon-like peptide 1 (GLP-1) acutely suppress appetite. The short chain fatty acid (SCFA) receptor, free fatty acid receptor 2 (FFA2) is present on colonic enteroendocrine L cells, and a role has been suggested for SCFAs in appetite regulation. Here, we characterise the in vitro and in vivo effects of colonic propionate on PYY and GLP-1 release in rodents, and investigate the role of FFA2 in mediating these effects using FFA2 knockout mice.

Methods: We used Wistar rats, C57BL6 mice and free fatty acid receptor 2 knockout (FFA(-/-)) mice on a C57BL6 background to explore the impact of the SCFA propionate on PYY and GLP-1 release. Isolated colonic crypt cultures were used to assess the effects of propionate on gut hormone release in vitro. We subsequently developed an in vivo technique to assess gut hormone release into the portal vein following colonic infusion of propionate.

Results: Propionate stimulated the secretion of both PYY and GLP-1 from wild-type primary murine colonic crypt cultures. This effect was significantly attenuated in cultures from FFA2(-/-) mice. Intra-colonic infusion of propionate elevated PYY and GLP-1 levels in jugular vein plasma in rats and in portal vein plasma in both rats and mice. However, propionate did not significantly stimulate gut hormone release in FFA2(-/-) mice.

Conclusions: Intra-colonic administration of propionate stimulates the concurrent release of both GLP-1 and PYY in rats and mice. These data demonstrate that FFA2 deficiency impairs SCFA-induced gut hormone secretion both in vitro and in vivo.

Figures

Figure 1
Figure 1
The effect of propionate on GLP-1 and PYY secretion from primary murine L cells. Mixed primary colonic cultures were incubated with propionate (1–50 mmol l−1) (a, c). At a concentration of 50 mmol l−1, NaCl had no effect on gut hormone release (b, d). GLP-1 and PYY secretion in each well was expressed as a percentage of total GLP-1 or PYY contained within the well and normalised to the basal secretion measured in parallel within the same experiment. Data represent means±s.e.m. (n=6–24 wells). Significance is shown relative to basal secretion (0 mmol l−1) (a, c) or to the iso-osmotic NaCl control (b, d) using one-way ANOVA (a, F=11.73, P<0.0001; b, F=7.931, P=0.0022; c, F=24.75, P<0.0001; d, F=25.99, P<0.0001) with a Bonferroni post hoc test (*P<0.05, **P<0.01, ***P<0.001).
Figure 2
Figure 2
Intra-colonic administration of propionate increases jugular and portal vein plasma gut hormone concentrations in male Wistar rats. Blood samples were collected over a 60-min period, via a jugular vein cannula (a, c), and at t=15 min from the portal vein (b, d), following an intra-colonic injection (2.5 ml) of 180 mmol l−1 propionate or saline (matched for pH and sodium content) in isoflurane-anaesthetised rats. Data represent means±s.e.m. (n=10–14 per group). Significance was determined using two-way ANOVA (a, c) or unpaired t-test (b, d) (*P<0.05).
Figure 3
Figure 3
Propionate-induced PYY and GLP-1 secretion is attenuated in FFA2−/− primary murine L cells. Primary colonic cultures from FFA2 knockout (−/−) or WT (+/+) littermates were incubated with or without propionate (50 mmol l−1). PYY (a) and GLP-1 (b) secretion in each well was expressed as a percentage of total PYY or GLP-1 contained within the well and normalised to the basal secretion (0 mmol l−1) measured in parallel within the same experiment. Data represent means±s.e.m. (n=22–38 wells). Significance is shown relative to basal secretion using one-way ANOVA (a, F=19.31, P<0.0001; b, F=8.816, P<0.0001) with a Bonferroni post hoc test (**P<0.01; ***P<0.001).
Figure 4
Figure 4
Intra-colonic administration of propionate increases portal vein plasma PYY and GLP-1 levels in vivo in mice via an FFA2-dependent mechanism. Blood samples were collected from the portal vein 5 min after an intra-colonic injection (300 μl) of propionate (180 mmol l−1) or saline (matched for pH and sodium content) in isoflurane-anaesthetised FFA2 knockout (−/−) mice or WT littermates (+/+). Data represent means±s.e.m. (n=5–7 per group). Significance was determined using one-way ANOVA (a, F=2.457, P=0.094; b, F=2.660, P=0.076) with a Bonferroni post hoc test (*P<0.05).

References

    1. Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. Nature. 2006;444:854–859.
    1. le Roux CW, Aylwin SJ, Batterham RL, Borg CM, Coyle F, Prasad V, et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243:108–114.
    1. le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246:780–785.
    1. Diakogiannaki E, Gribble FM, Reimann F. Nutrient detection by incretin hormone secreting cells. Physiol Behav. 2012;106:387–393.
    1. Eissele R, Goke R, Willemer S, Harthus HP, Vermeer H, Arnold R, et al. Glucagon-like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and man. Eur J Clin Invest. 1992;22:283–291.
    1. Mortensen PB, Clausen MR. Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease. Scand J Gastroenterol Suppl. 1996;216:132–148.
    1. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun. 2013;4:1829.
    1. Nøhr MK, Pedersen MH, Gille A, Egerod KL, Engelstoft MS, Husted AS, et al. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology. 2013;154:3552–3564.
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–573.
    1. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes. 2012;61:364–371.
    1. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278:25481–25489.
    1. Liou AP, Paziuk M, Luevano JM, Jr., Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5:178ra41.
    1. Morrison DJ, Mackay WG, Edwards CA, Preston T, Dodson B, Weaver LT. Butyrate production from oligofructose fermentation by the human faecal flora: what is the contribution of extracellular acetate and lactate. Br J Nutr. 2006;96:570–577.
    1. Reimann F, Habib AM, Tolhurst G, Parker HE, Rogers GJ, Gribble FM. Glucose sensing in L cells: a primary cell study. Cell Metab. 2008;8:532–539.
    1. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286.
    1. Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br J Nutr. 2004;92:521–526.
    1. Delmée E, Cani PD, Gual G, Knauf C, Burcelin R, Maton N, et al. Relation between colonic proglucagon expression and metabolic response to oligofructose in high fat diet-fed mice. Life Sci. 2006;79:1007–1013.
    1. Adrian TE, Ferri GL, Bacarese-Hamilton AJ, Fuessl HS, Polak JM, Bloom SR. Human distribution and release of a putative new gut hormone, peptide YY. Gastroenterology. 1985;89:1070–1077.
    1. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet. 1987;2:1300–1304.
    1. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–2340.
    1. Karaki S, Mitsui R, Hayashi H, Kato I, Sugiya H, Iwanaga T, et al. Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 2006;324:353–360.
    1. Karaki S, Tazoe H, Hayashi H, Kashiwabara H, Tooyama K, Suzuki Y, et al. Expression of the short-chain fatty acid receptor, GPR43, in the human colon. J Mol Histol. 2008;39:135–142.
    1. Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. 2003;278:11312–11319.
    1. Anini Y, Fu-Cheng X, Cuber JC, Kervran A, Chariot J, Roz C. Comparison of the postprandial release of peptide YY and proglucagon-derived peptides in the rat. Pflugers Arch. 1999;438:299–306.
    1. Plaisancié P, Dumoulin V, Chayvialle JA, Cuber JC. Luminal glucagon-like peptide-1(7-36) amide-releasing factors in the isolated vascularly perfused rat colon. J Endocrinol. 1995;145:521–526.
    1. Cherbut C, Ferrier L, Roze C, Anini Y, Blottiere H, Lecannu G, et al. Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am J Physiol. 1998;275:G1415–G1422.
    1. Fu-Cheng X, Anini Y, Chariot J, Voisin T, Galmiche JP, Rozé C. Peptide YY release after intraduodenal, intraileal, and intracolonic administration of nutrients in rats. Pflugers Arch. 1995;431:66–75.
    1. Cani PD, Montoya ML, Neyrinck AM, Delzenne NM, Lambert DM. Potential modulation of plasma ghrelin and glucagon-like peptide-1 by anorexigenic cannabinoid compounds, SR141716A (rimonabant) and oleoylethanolamide. Br J Nutr. 2004;92:757–761.
    1. Saeidi N, Meoli L, Nestoridi E, Gupta NK, Kvas S, Kucharczyk J, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341:406–410.
    1. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214.
    1. Bjursell M, Admyre T, Goransson M, Marley AE, Smith DM, Oscarsson J, et al. Improved glucose control and reduced body fat mass in free fatty acid receptor 2-deficient mice fed a high-fat diet. Am J Physiol Endocrinol Metab. 2011;300:E211–E220.
    1. Cani PD, Hoste S, Guiot Y, Delzenne NM. Dietary non-digestible carbohydrates promote L-cell differentiation in the proximal colon of rats. Br J Nutr. 2007;98:32–37.
    1. Petersen N, Reimann F, Bartfeld S, Farin HF, Ringnalda FC, Vries RG, et al. Generation of L cells in mouse and human small intestine organoids. Diabetes. 2014;63:410–420.

Source: PubMed

3
Subskrybuj