DOI QR코드

DOI QR Code

Perspectives on the therapeutic potential of short-chain fatty acid receptors

  • Kim, Sunhong (Targeted Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology) ;
  • Kim, Jeong-Hoon (Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology) ;
  • Park, Bi Oh (Targeted Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology) ;
  • Kwak, Young Shin (College of Pharmacy, Korea University)
  • Received : 2013.12.03
  • Accepted : 2013.01.13
  • Published : 2014.03.31

Abstract

There is rapidly growing interest in the human microbiome because of its implication in metabolic disorders and inflammatory diseases. Consequently, understanding the biology of short chain fatty acids and their receptors has become very important for identifying novel therapeutic avenues. GPR41 and GPR43 have been recognized as the cognate receptors for SCFAs and their roles in metabolism and inflammation have drawn much attention in recent years. GPR43 is highly expressed on immune cells and has been suggested to play a role in inflammatory diseases such as inflammatory bowel disease. Both GPR41 and GPR43 have been implicated in diabetes and obesity via the regulation of adipose tissue and gastrointestinal hormones. So far, many studies have provided contradictory results, and therefore further research is required to validate these receptors as drug targets. We will also discuss the synthetic modulators of GPR41 and GPR43 that are critical to understanding the functions of these receptors.

Keywords

INTRODUCTION

G-protein-coupled receptors (GPCRs) comprise a large family of seven-transmembrane receptors that convey various extracellular stimuli such as light, odorants, cytokines and hormones to a cascade of intracellular signaling. There are ∼800 predicted human GPCRs of which over 300 are non-olfactory receptors that are involved in various physiological processes (1,2) and can be categorized into six subfamilies (Class A-F) based on their conserved residues and ligands (3,4). The importance of GPCRs is emphasized by the fact that 27% of FDA-approved drugs target GPCRs (5), although only a minor portion of these non-olfactory receptors are exploited as therapeutic targets. GPCRs activate heterotrimeric G proteins, where the α and βγ subunits are responsible for signaling, and independent from the G protein signaling they also recruit β-arrestins to be desensitized and transduce a signal.

Fatty acids are categorized by the number of their carbons. Short chain fatty acids (SCFAs) refer to carboxylic acids with aliphatic tails of less than 6 carbons such as formic, acetic, propionic, butyric, isobutyric, valeric, isovaleric, and 2-methylbutyric acids (6). SCFAs are typically synthesized in vivo, whereas longer chain fatty acids are often derived from food sources. Among them, acetate, propionate, and butyrate consist of over 90% of the SCFAs in the colon and are largely produced by multiple anaerobic bacteria in the human gut (7). SCFAs have been regarded as an energy source for gut epithelium and peripheral tissues, as well as a regulator of metabolism and inflammation (8). A decade ago, it was found that two GPCRs, GPR41 (FFAR3/FFA3) and GPR43 (FFAR2/FFA2), can sense these fatty acids (9-11). These receptors are expressed not only in the gut endocrine cells in the vicinity of SCFA production, but also at multiple other sites such as adipose tissue, pancreatic islets, and immune cells (9-13). Because of the relatively recent deorphanization of GPR41 and GPR43, more and more studies have been emerging that unveil the roles of these receptors, and their SCFA ligands, in human physiology. This review will focus on the biological functions of GPR41 and GPR43 as relevant to drug targeting and outline the current status of drug discovery with these receptors.

 

BIOLOGY OF SCFA RECEPTORS

GPR41 and GPR43 were first identified, along with GPR40, as constituents of a novel GPCR gene cluster spanning about 100kb in human chromosome 19q13.1 (14). They were later found to be activated by SCFAs, which showed a distinct structure-activity relationship toward these two receptors. The order of potency for GPR43 was propionate = acetate > butyrate > valerate = formate, while for GPR41 it was propionate = butyrate = valerate > acetate = formate (9-11). However, these ranked orders of potency were later found to be not conserved in other species (15). Although both receptors respond to the same group of ligands, GPR41 and GPR43 are coupled to a different set of downstream signaling cascades. GPR41 selectively couples with Gαi, which inhibits adenylyl cyclase, so that its activation by SCFAs decreases the intracellular concentration of cyclic AMP (cAMP) (9,10). Acetate and propionate activate GPR43 and in turn, induce both Gαi and Gαq, which decreases cAMP and increases cytoplasmic calcium ions (9-11). In addition, our group recently showed that Nuclear Factor κB (NFκB) can be regulated by the GPR43-β-arrestin 2 pathway (16), whereas, as far as we know, there is no report that has linked GPR41 to β-arrestins.

GPR41 was initially found to be broadly expressed in various tissues, including the pancreas, spleen and adipose tissue. Subsequently, it was found that SCFAs stimulate the production of the hormone leptin in mouse white adipose tissue cells via GPR41 (17). However, several groups were unable to detect GPR41 in mouse adipose tissue or in a differentiated adipocyte cell line, but instead found GPR43 to be highly expressed in those cells (18-21). In addition, GPR41 is present in a variety of enteroendocrine cells and enteric neurons with varying degrees of expression level and is implicated in the release of gastrointestinal hormones (22,23). Like the controversy over its expression in adipose tissue, one group reported that GPR41-knockout (KO) mice had significantly less fat than wild-type mice (23), while another group later showed no significant difference between the fat content of wild type and GPR41 KO mice (24). In addition, Zaibi and co-workers disclosed their unpublished results, which found that male GPR41 KO mice were obese rather than lean, supporting their prediction that GPR41 knockout mice would have reduced leptin levels because of decreased expression of GPR43 in adipose tissue and subsequently, reduced stimulation of leptin secretion by plasma SCFA (19). These discrepancies may arise from the different genetic backgrounds or rearing conditions in the various experiments, which would affect the gut microbiota. Finally, a pair of recent papers revealed an intriguing physiological function of GPR41. In the fed state, SCFAs and ketones modulate the sympathetic nervous system directly through the GPR41-Gβγ-PLCβ-ERK axis to contribute to sympathetic activation (21,25). Taken together, GPR41 agonists might have a beneficial effect on the treatment of obesity and other metabolic diseases. To clarify the in vivo functions of GPR41, careful examination of KO mice and highly specific small molecule modulators would be needed.

GPR43 is expressed in the adipose tissue, intestine, and especially immune cells such as peripheral blood mononuclear cells (PBMC), eosinophils, and neutrophils (9-11,22,26-28). For the last decade, it has been quite well established that SCFAs induce neutrophil chemotaxis via GPR43 in p38 and Akt-dependent manners (10,28-30). Intriguingly, more recent studies showed contradictory results that GPR43 KO mice had either exacerbated or reduced inflammation in a colitis model (28,29,31,32). Although a link between GPR43 and inflammatory bowel disease (IBD) (and possibly other inflammatory diseases) seems to be firm, it remains to be resolved whether an agonist or antagonist of GPR43 would be the correct treatment for this disease.

The plasma levels of free fatty acids are usually increased in metabolic diseases such as obesity and type II diabetes, resulting in insulin resistance and lipid deposition (33,34). The downregulation of lipolysis in the adipose tissue could therefore be a plausible pharmacological strategy for improving insulin sensitivity. The data showing that GPR43 is expressed in adipose tissue and differentiated adipocytes raised the possibility of the involvement of GPR43 in lipid metabolism. Indeed, SCFAs enhanced adipogenesis and inhibited lipolysis, whereas knockdown or knockout of GPR43 abolished these effects (18,35). In agreement with these results, a synthetic GPR43 agonist attenuated lipolysis (36-38). As was the case for GPR41, studies of GPR43 KO yielded conflicting phenotypes. When mice deficient for GPR43 were fed a high fat diet, the mice displayed lower body fat mass, improved glucose control, lower levels of plasma lipids, and decreased inflammation in white adipose tissue compared to the wild type, probably due to the elevated body temperature and subsequently increased energy expenditure of the GPR43 deficient mice (39). However, Tsujimoto and co-workers demonstrated that GPR43 KO mice were obese on a normal diet, whereas mice overexpressing GPR43 in adipose tissue were lean even when fed a high-fat diet (20). In that paper, they hypothesized that this discrepancy may result from the differences in the genetic backgrounds of the mice used in the different studies. It is the latter study that is more likely, given that GPR43 and SCFAs promote the release of peptide YY and glucagon-like peptide-1, which is an anorexic hormone and incretin hormone, respectively (26,27,40,41). Taken together, these data strongly indicate that GPR43 agonists have potential as therapeutics for the treatment of type II diabetes and obesity.

 

COMPOUNDS TARGETING SCFA RECEPTORS: AGONISTS OR ANTAGONISTS?

As shown in Table 1, early discovery efforts produced a series of allosteric agonists by Amgen (36,42,43) and orthosteric agonists generated by Euroscreen SA (44). The series of allosteric compounds from Amgen were completely selective for GPR43 over GPR41 (S. Kim, unpublished result). These compounds alone produced a full agonistic response as compared to SCFAs and showed similar potency toward human GPR43 and mouse GPR43, and acted in a positively cooperative fashion with SCFAs (36,42,43). In addition, these phenylacetamide compounds have been shown to inhibit lipolysis in differentiated adipocytes and lower free fatty acid levels in mice serum (45,46), which is consistent with the previous data from siRNA treated and KO mice. Euroscreen SA developed many synthetic orthosteric ligands with better potency that displayed similar effects such as increased glucose uptake, decreased lipolysis, increased GLP-1 level, improved oral glucose tolerance test, and reduced TNFα production (37,47,48). An antagonist developed by Galapagos for the treatment of psoriasis was announced as an orally available small molecule that reduced migration of neutrophils in rodent studies (49). Presently, this is the only compound in clinical phase among the different modulators of GPR43. In contrast to GPR43, the development of GPR41 modulators has been less transparent. A GPR41-selective agonist and antagonist were developed by Arena pharmaceuticals (50). However, the patent did not disclose the potency of either of these compounds.

Table 1.Selective agonists and antagonists of GPR41 and GPR43

 

CONCLUDING REMARKS

Although there is a variety of evidence indicating that GPR41 and GPR43 are involved in disease-relevant physiological processes such as lipolysis, adipogenesis, hormone secretion, and inflammation, it remains to be elucidated whether these receptors are valuable drug targets. It is also currently under debate which direction of modulation - agonism vs. antagonism - would yield therapeutic benefits. The question of whether GPR41 and GPR43 are druggable targets is a difficult one. Answers to this would be best provided by more research on the biology of GPR41 and GPR43and the selective potent compounds targeting these receptors.

References

  1. Fredriksson, R., Lagerstrom, M. C., Lundin, L. G. and Schioth, H. B. (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol. Pharmacol. 63, 1256-1272. https://doi.org/10.1124/mol.63.6.1256
  2. Lagerstrom, M. C. and Schioth, H. B. (2008) Structural diversity of G protein-coupled receptors and significance for drug discovery. Nat. Rev. Drug Discov. 7, 339-357. https://doi.org/10.1038/nrd2518
  3. Kolakowski, L. F., Jr. (1994) GCRDb: a G-protein-coupled receptor database. Receptors Channels 2, 1-7.
  4. Bockaert, J. and Pin, J. P. (1999) Molecular tinkering of G protein-coupled receptors: an evolutionary success. EMBO J. 18, 1723-1729. https://doi.org/10.1093/emboj/18.7.1723
  5. Overington, J. P., Al-Lazikani, B. and Hopkins, A. L. (2006) How many drug targets are there? Nat. Rev. Drug Discov. 5, 993-996. https://doi.org/10.1038/nrd2199
  6. Bergman, E. N. (1990) Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol. Rev. 70, 567-590. https://doi.org/10.1152/physrev.1990.70.2.567
  7. Mortensen, P. B. and Clausen, M. R. (1996) Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease. Scand. J. Gastroenterol. Suppl. 216, 132-148.
  8. den Besten, G., van Eunen, K., Groen, A. K., Venema, K., Reijngoud, D. J. and Bakker, B. M. (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, 2325-2340. https://doi.org/10.1194/jlr.R036012
  9. Brown, A. J., Goldsworthy, S. M., Barnes, A. A., Eilert, M. M., Tcheang, L., Daniels, D., Muir, A. I., Wigglesworth, M. J., Kinghorn, I., Fraser, N. J., Pike, N. B., Strum, J. C., Steplewski, K. M., Murdock, P. R., Holder, J. C., Marshall, F. H., Szekeres, P. G., Wilson, S., Ignar, D. M., Foord, S. M., Wise, A. and Dowell, S. J. (2003) 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. https://doi.org/10.1074/jbc.M211609200
  10. Le Poul, E., Loison, C., Struyf, S., Springael, J. Y., Lannoy, V., Decobecq, M. E., Brezillon, S., Dupriez, V., Vassart, G., Van Damme, J., Parmentier, M. andDetheux, M. (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 278, 25481-25489. https://doi.org/10.1074/jbc.M301403200
  11. Nilsson, N. E., Kotarsky, K., Owman, C. and Olde, B. (2003) Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem. Biophys. Res. Commun. 303, 1047-1052. https://doi.org/10.1016/S0006-291X(03)00488-1
  12. Nakajima, T., Iikura, M., Okayama, Y., Matsumoto, K., Uchiyama, C., Shirakawa, T., Yang, X., Adra, C. N., Hirai, K. and Saito, H. (2004) Identification of granulocyte subtype-selective receptors and ion channels by using a high-density oligonucleotide probe array. J. Allergy Clin. Immunol. 113, 528-535. https://doi.org/10.1016/j.jaci.2003.12.036
  13. Senga, T., Iwamoto, S., Yoshida, T., Yokota, T., Adachi, K., Azuma, E., Hamaguchi, M. and Iwamoto, T. (2003) LSSIG is a novel murine leukocyte-specific GPCR that is induced by the activation of STAT3. Blood 101, 1185-1187. https://doi.org/10.1182/blood-2002-06-1881
  14. Sawzdargo, M., George, S. R., Nguyen, T., Xu, S., Kolakowski, L. F. and O'Dowd, B. F. (1997) A cluster of four novel human G protein-coupled receptor genes occurring in close proximity to CD22 gene on chromosome 19q13.1. Biochem. Biophys. Res. Commun. 239, 543-547. https://doi.org/10.1006/bbrc.1997.7513
  15. Hudson, B. D., Tikhonova, I. G., Pandey, S. K., Ulven, T. and Milligan, G. (2012) Extracellular ionic locks determine variation in constitutive activity and ligand potency between species orthologs of the free fatty acid receptors FFA2 and FFA3. J. Biol. Chem. 287, 41195-41209. https://doi.org/10.1074/jbc.M112.396259
  16. Lee, S. U., In, H. J., Kwon, M. S., Park, B. O., Jo, M., Kim, M. O., Cho, S., Lee, S., Lee, H. J., Kwak, Y. S. and Kim, S. (2013) beta-Arrestin 2 Mediates G Protein-Coupled Receptor 43 Signals to Nuclear Factor-kappaB. Biol. Pharm. Bull. 36, 1754-1759. https://doi.org/10.1248/bpb.b13-00312
  17. Xiong, Y., Miyamoto, N., Shibata, K., Valasek, M. A., Motoike, T., Kedzierski, R. M. and Yanagisawa, M. (2004) Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl. Acad. Sci. U. S. A. 101, 1045-1050. https://doi.org/10.1073/pnas.2637002100
  18. Hong, Y. H.,Nishimura, Y., Hishikawa, D., Tsuzuki, H., Miyahara, H., Gotoh, C., Choi, K. C., Feng, D. D., Chen, C., Lee, H. G., Katoh, K., Roh, S. G. and Sasaki, S. (2005) Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology 146, 5092-5099. https://doi.org/10.1210/en.2005-0545
  19. Zaibi, M. S., Stocker, C. J., O'Dowd, J., Davies, A., Bellahcene, M., Cawthorne, M. A., Brown, A. J., Smith, D. M. and Arch, J. R. (2010) Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett. 584, 2381-2386. https://doi.org/10.1016/j.febslet.2010.04.027
  20. Kimura, I., Ozawa, K., Inoue, D., Imamura, T., Kimura, K., Maeda, T., Terasawa, K., Kashihara, D., Hirano, K., Tani, T., Takahashi, T., Miyauchi, S., Shioi, G., Inoue, H. and Tsujimoto, G. (2013) The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 4, 1829. https://doi.org/10.1038/ncomms2852
  21. Kimura, I., Inoue, D., Maeda, T., Hara, T., Ichimura, A., Miyauchi, S., Kobayashi, M., Hirasawa, A. and Tsujimoto, G. (2011) Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl. Acad. Sci. U. S. A. 108, 8030-8035. https://doi.org/10.1073/pnas.1016088108
  22. Nohr, M. K., Pedersen, M. H., Gille, A., Egerod, K. L., Engelstoft, M. S., Husted, A. S., Sichlau, R. M., Grunddal, K. V., Poulsen, S. S., Han, S., Jones, R. M., Offermanns, S. and Schwartz, T. W. (2013) 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 154, 3552-3564. https://doi.org/10.1210/en.2013-1142
  23. Samuel, B. S., Shaito, A., Motoike, T., Rey, F. E., Backhed, F., Manchester, J. K., Hammer, R. E., Williams, S. C., Crowley, J., Yanagisawa, M. and Gordon, J. I. (2008) 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. U. S. A. 105, 16767-16772. https://doi.org/10.1073/pnas.0808567105
  24. Lin, H. V., Frassetto, A., Kowalik, E. J., Jr., Nawrocki, A. R., Lu, M. M., Kosinski, J. R., Hubert, J. A., Szeto, D., Yao, X., Forrest, G. and Marsh, D. J. (2012) Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One 7, e35240. https://doi.org/10.1371/journal.pone.0035240
  25. Inoue, D., Kimura, I., Wakabayashi, M.,Tsumoto, H., Ozawa, K., Hara, T., Takei, Y., Hirasawa, A., Ishihama, Y. and Tsujimoto, G. (2012) Short-chain fatty acid receptor GPR41-mediated activation of sympathetic neurons involves synapsin 2b phosphorylation. FEBS Lett. 586, 1547-1554. https://doi.org/10.1016/j.febslet.2012.04.021
  26. Karaki, S., Mitsui, R., Hayashi, H., Kato, I., Sugiya, H., Iwanaga, T., Furness, J. B. and Kuwahara, A. (2006) Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 324, 353-360. https://doi.org/10.1007/s00441-005-0140-x
  27. Karaki, S., Tazoe, H., Hayashi, H., Kashiwabara, H., Tooyama, K., Suzuki, Y. and Kuwahara, A. (2008) Expression of the short-chain fatty acid receptor, GPR43, in the human colon. J. Mol. Histol. 39, 135-142. https://doi.org/10.1007/s10735-007-9145-y
  28. Maslowski, K. M., Vieira, A. T., Ng, A., Kranich, J., Sierro, F., Yu, D., Schilter, H. C., Rolph, M. S., Mackay, F., Artis, D., Xavier, R. J., Teixeira, M. M. and Mackay, C. R. (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282-1286. https://doi.org/10.1038/nature08530
  29. Sina, C., Gavrilova, O., Forster, M., Till, A., Derer, S., Hildebrand, F., Raabe, B., Chalaris, A., Scheller, J., Rehmann, A., Franke, A., Ott, S., Hasler, R., Nikolaus, S., Folsch, U. R., Rose-John, S., Jiang, H. P., Li, J., Schreiber, S. and Rosenstiel, P. (2009) G protein-coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation. J. Immunol. 183, 7514-7522. https://doi.org/10.4049/jimmunol.0900063
  30. Vinolo, M. A., Ferguson, G. J., Kulkarni, S., Damoulakis, G., Anderson, K., Bohlooly, Y. M., Stephens, L., Hawkins, P. T. and Curi, R. (2011) SCFAs induce mouse neutrophil chemotaxis through the GPR43 receptor. PLoS One 6, e21205. https://doi.org/10.1371/journal.pone.0021205
  31. Kim, M. H., Kang, S. G., Park, J. H., Yanagisawa, M. and Kim, C. H. (2013) Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 145, 396-406 e391-310. https://doi.org/10.1053/j.gastro.2013.04.056
  32. Smith, P. M., Howitt, M. R., Panikov, N., Michaud, M., Gallini, C. A., Bohlooly, Y. M., Glickman, J. N. and Garrett, W. S. (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569-573. https://doi.org/10.1126/science.1241165
  33. Haber, E. P., Ximenes, H. M., Procopio, J., Carvalho, C. R., Curi, R. and Carpinelli, A. R. (2003) Pleiotropic effects of fatty acids on pancreatic beta-cells. J. Cell Physiol. 194, 1-12. https://doi.org/10.1002/jcp.10187
  34. Reaven, G. M., Hollenbeck, C., Jeng, C. Y., Wu, M. S. and Chen, Y. D. (1988) Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes 37, 1020-1024. https://doi.org/10.2337/diab.37.8.1020
  35. Ge, H., Li, X., Weiszmann, J., Wang, P., Baribault, H., Chen, J. L., Tian, H. and Li, Y. (2008) 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. https://doi.org/10.1210/en.2008-0059
  36. Lee, T., Schwandner, R., Swaminath, G., Weiszmann, J., Cardozo, M., Greenberg, J., Jaeckel, P., Ge, H., Wang, Y., Jiao, X., Liu, J., Kayser, F., Tian, H. and Li, Y. (2008) Identification and functional characterization of allosteric agonists for the G protein-coupled receptor FFA2. Mol. Pharmacol. 74, 1599-1609. https://doi.org/10.1124/mol.108.049536
  37. EUROSCREEN (2010) COMPOUNDS, PHARMACEUTICAL COMPOSITION AND METHODS FOR USE IN TREATING METABOLIC DISORDERS. WIPO WO 2010/066682 A1.
  38. Hudson, B. D., Due-Hansen, M. E., Christiansen, E., Hansen, A. M., Mackenzie, A. E., Murdoch, H., Pandey, S. K., Ward, R. J., Marquez, R., Tikhonova, I. G., Ulven, T. and Milligan, G. (2013) Defining the molecular basis for the first potent and selective orthosteric agonists of the FFA2 free fatty acid receptor. J. Biol. Chem. 288, 17296-17312. https://doi.org/10.1074/jbc.M113.455337
  39. Bjursell, M., Admyre, T., Goransson, M., Marley, A. E., Smith, D. M., Oscarsson, J. and Bohlooly, Y. M. (2011) 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. 300, E211-220. https://doi.org/10.1152/ajpendo.00229.2010
  40. Karra, E. and Batterham, R. L. (2010) The role of gut hormones in the regulation of body weight and energy homeostasis. Mol. Cell. Endocrinol. 316, 120-128. https://doi.org/10.1016/j.mce.2009.06.010
  41. Tolhurst, G., Heffron, H., Lam,Y. S., Parker, H. E., Habib, A. M., Diakogiannaki, E., Cameron, J., Grosse, J., Reimann, F. and Gribble, F. M. (2012) Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364-371. https://doi.org/10.2337/db11-1019
  42. Wang, Y., Jiao, X., Kayser, F., Liu, J., Wang, Z., Wanska, M., Greenberg, J., Weiszmann, J., Ge, H., Tian, H., Wong, S., Schwandner, R., Lee, T. and Li, Y. (2010) The first synthetic agonists of FFA2: Discovery and SAR of phenylacetamides as allosteric modulators. Bioorg. Med. Chem. Lett. 20, 493-498. https://doi.org/10.1016/j.bmcl.2009.11.112
  43. Swaminath, G., Jaeckel, P., Guo, Q., Cardozo, M., Weiszmann, J., Lindberg, R., Wang, Y., Schwandner, R. and Li, Y. (2011) Mutational analysis of G-protein coupled receptor--FFA2. Biochem. Biophys. Res. Commun. 405, 122-127. https://doi.org/10.1016/j.bbrc.2010.12.139
  44. Bernard, J., Hartiel, A. F., Brantis, C., Hoveyda, H. and Fraser, G. (2009) Identification of a small-molecule GPR43 agonist that Increases glucose uptake and inhibits lipolysis in adipocytes. American Diabetes Association Annual Meeting, New Orleans, LA, USA 69, Abs1362p.
  45. Lee, T., Schwandner, R., Swaminath, G., Weiszmann, J., Cardozo, M., Greenberg, J., Jaeckel, P., Ge, H., Wang, Y., Jiao, X., Liu, J., Kayser, F., Tian, H. and Li, Y. (2008) Identification and functional characterization ofallosteric agonists for the G protein-coupled receptor FFA2. Mol. Pharmacol. 74, 1599-1609. https://doi.org/10.1124/mol.108.049536
  46. Wang, Y., Jiao, X., Kayser, F., Liu, J., Wang, Z., Wanska, M., Greenberg, J., Weiszmann, J., Ge, H., Tian, H., Wong, S., Schwandner, R., Lee, T. and Li, Y. (2010) The first synthetic agonists of FFA2: Discovery and SAR of phenylacetamides as allosteric modulators. Bioorg. Med. Chem. Lett. 20, 493-498. https://doi.org/10.1016/j.bmcl.2009.11.112
  47. EUROSCREEN (2011) PYRROLIDINE OR THIAZOLIDINE CARBOXYLIC ACID DERIVATIVES, PHARMACEUTICAL COMPOSITION AND METHODS FOR USE IN TREATING METABOLIC DISORDERS AS AGONISTS OF G-PROTEIN COUPLED RECEPTOR 43 (GPR43). WIPO WO2011/073376A1.
  48. EUROSCREEN (2011) NOVEL COMPOUNDS, METHOD FOR USE THEM AND PHARMACEUTICAL COMPOSITION CONTAINING THEM. WIPO WO 2011/151436 A2.
  49. Saniere, L. R. M., Pizzonero, M. R., Triballeu, N., Vandeghinste, N. E. R., De Vos, S. I. J., Brys, R. C. X. and Pourbaix-Lebraly, C. D. (2012) AZETIDINE DERIVATIVES USEFUL FOR THE TREATMENT OF METABOLIC AND INFLAMMATORY DISEASES. WIPO WO 2012/098033A1.
  50. Leonard, J. N., Chu, Z. L., Bruce, M. A. and Boatman, P. D. (2006) GPR41 and modulators thereof for the treatment of insulin-related disorders. WIPO WO2006/052566 A2.

Cited by

  1. Complex Pharmacology of Free Fatty Acid Receptors vol.117, pp.1, 2017, https://doi.org/10.1021/acs.chemrev.6b00056
  2. Diet as a Trigger or Therapy for Inflammatory Bowel Diseases vol.152, pp.2, 2017, https://doi.org/10.1053/j.gastro.2016.10.019
  3. Advanced Application of Porcine Intramuscular Adipocytes for Evaluating Anti-Adipogenic and Anti-Inflammatory Activities of Immunobiotics vol.10, pp.3, 2015, https://doi.org/10.1371/journal.pone.0119644
  4. The role of adipose tissue immune cells in obesity and low-grade inflammation vol.222, pp.3, 2014, https://doi.org/10.1530/JOE-14-0283
  5. Western diet induces a shift in microbiota composition enhancing susceptibility to Adherent-Invasive E. coli infection and intestinal inflammation. vol.6, pp.1, 2016, https://doi.org/10.1038/srep19032
  6. Gut as an emerging organ for the treatment of diabetes: focus on mechanism of action of bariatric and endoscopic interventions vol.237, pp.1, 2018, https://doi.org/10.1530/JOE-17-0438
  7. The Role of Metabolite-Sensing G Protein-Coupled Receptors in Inflammation and Metabolic Disease vol.29, pp.3, 2018, https://doi.org/10.1089/ars.2017.7168