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:

Targeting bile-acid signalling for metabolic diseases

Key Points

  • Hepatic synthesis of bile acids is the primary pathway for cholesterol catabolism. The cholesterol 7α-hydroxylase enzyme (encoded by CYP7A1) represents the rate-limiting step of the multi-enzymatic bile-acid biosynthetic pathway.

  • Bile acids play a crucial role in dietary lipid digestion and absorption, and also act as versatile signalling molecules through the activation of the nuclear hormone receptor farnesoid X receptor-α (FXR-α) and the recently identified G-protein-coupled receptor TGR5.

  • Bile-acid-mediated activation of FXR-α-signalling pathways regulate the enterohepatic recycling of bile acids, protect against their accumulation in the liver and inhibit their own biosynthesis.

  • Through their endocrine function, bile acids also activate TGR5 signalling pathways in multiple cells, through which they control immune function, liver and gall-bladder physiology and glucose and energy homeostasis.

  • The development of TGR5 agonists could have benefits to combat many aspects of the metabolic syndrome, whereas FXR-α agonists could hold promise for reducing hypertriglyceridaemia and modulating glucose metabolism. Pharmaceutical activation of these bile-acid-signalling pathways is therefore a novel way to improve metabolism.

Abstract

Bile acids are increasingly being appreciated as complex metabolic integrators and signalling factors and not just as lipid solubilizers and simple regulators of bile-acid homeostasis. It is therefore not surprising that a number of bile-acid-activated signalling pathways have become attractive therapeutic targets for metabolic disorders. Here, we review how the signalling functions of bile acids can be exploited in the development of drugs for obesity, type 2 diabetes, hypertriglyceridaemia and atherosclerosis, as well as other associated chronic diseases such as non-alcoholic steatohepatitis.

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

Figure 1: Bile-acid synthesis pathways.
Figure 2: Structure and hydrophobic/hydrophilic profile of bile acids.
Figure 3: Overview of the bile-acid transport system.
Figure 4: FXR-mediated genomic actions of bile acids.
Figure 5: TGR5: a dedicated membrane bile-acid receptor.
Figure 6: A model for the TGR5 ligand binding pocket.

Similar content being viewed by others

References

  1. Houten, S. M. & Auwerx, J. The enterohepatic nuclear receptors are major regulators of the enterohepatic circulation of bile salts. Ann. Med. 36, 482–491 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Ho, K. J. Circadian distribution of bile acids in the enterohepatic circulatory system in rats. Am. J. Physiol. 230, 1331–1335 (1976).

    Article  CAS  PubMed  Google Scholar 

  3. Engelking, L. R., Dasher, C. A. & Hirschowitz, B. I. Within-day fluctuations in serum bile-acid concentrations among normal control subjects and patients with hepatic disease. Am. J. Clin. Pathol. 73, 196–201 (1980).

    Article  CAS  PubMed  Google Scholar 

  4. Everson, G. T. Steady-state kinetics of serum bile acids in healthy human subjects: single and dual isotope techniques using stable isotopes and mass spectrometry. J. Lipid Res. 28, 238–252 (1987).

    Article  CAS  PubMed  Google Scholar 

  5. Russell, D. W. The enzymes, regulation, and genetics of bile acid synthesis. Annu. Rev. Biochem. 72, 137–174 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Schwarz, M. et al. Disruption of cholesterol 7α-hydroxylase gene in mice. II. Bile acid deficiency is overcome by induction of oxysterol 7α-hydroxylase. J. Biol. Chem. 271, 18024–18031 (1996).

    Article  CAS  PubMed  Google Scholar 

  7. Ishibashi, S., Schwarz, M., Frykman, P. K., Herz, J. & Russell, D. W. Disruption of cholesterol 7α-hydroxylase gene in mice. I. Postnatal lethality reversed by bile acid and vitamin supplementation. J. Biol. Chem. 271, 18017–18023 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Beigneux, A., Hofmann, A. F. & Young, S. G. Human CYP7A1 deficiency: progress and enigmas. J. Clin. Invest. 110, 29–31 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pullinger, C. R. et al. Human cholesterol 7α-hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. J. Clin. Invest. 110, 109–117 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Salen, G. Cholestanol deposition in cerebrotendinous xanthomatosis. A possible mechanism. Ann. Intern. Med. 75, 843–851 (1971).

    Article  CAS  PubMed  Google Scholar 

  11. Salen, G. & Grundy, S. M. The metabolism of cholestanol, cholesterol, and bile acids in cerebrotendinous xanthomatosis. J. Clin. Invest. 52, 2822–2835 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Botham, K. M. & Boyd, G. S. The metabolism of chenodeoxycholic acid to β-muricholic acid in rat liver. Eur. J. Biochem. 134, 191–196 (1983).

    Article  CAS  PubMed  Google Scholar 

  13. Maruyama, T. et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem. Biophys. Res. Commun. 298, 714–719 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Kawamata, Y. et al. A G protein-coupled receptor responsive to bile acids. J. Biol. Chem. 278, 9435–9440 (2003). References 13 and 14 report the identification of TGR5, a GPCR dedicated to bile acids..

    Article  CAS  PubMed  Google Scholar 

  15. Makishima, M. et al. Identification of nuclear receptors for bile acids. Science 284, 1362–1365 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Parks, D. J. et al. Bile acids: natural ligands for orphan nuclear receptors. Science 284, 1365–1368 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Wang, H., Chen, J., Hollister, K., Sowers, L. C. & Forman, B. M. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol. Cell 3, 543–553 (1999). References15–17 identify bile acids as endogenous ligands of the nuclear receptor FXR.

    Article  CAS  PubMed  Google Scholar 

  18. Forman, B. M. et al. Identification of a nuclear receptor that is activated by Farnesol metabolites. Cell 81, 687–693 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Seol, W., Choi, H. S. & Moore, D. D. Isolation of proteins that interact specifically with retinoid X receptor: two novel orphan receptors. Mol. Endocrinol. 9, 72–85 (1995).

    CAS  PubMed  Google Scholar 

  20. Otte, K. et al. Identification of farnesoid X receptor beta as a novel mammalian nuclear receptor sensing lanosterol. Mol. Cell Biol. 23, 864–872 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Huber, R. M. et al. Generation of multiple farnesoid-X-receptor isoforms through the use of alternative promoters. Gene 290, 35–43 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Zhang, Y., Kast-Woelbern, H. R. & Edwards, P. A. Natural structural variants of the nuclear receptor farnesoid X receptor affect transcriptional activation. J. Biol. Chem. 278, 104–110 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Zhang, Y., Castellani, L. W., Sinal, C. J., Gonzalez, F. J. & Edwards, P. A. Peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) regulates triglyceride metabolism by activation of the nuclear receptor FXR. Genes Dev. 18, 157–169 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mi, L. Z. et al. Structural basis for bile acid binding and activation of the nuclear receptor FXR. Mol. Cell 11, 1093–1100 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Downes, M. et al. A chemical, genetic, and structural analysis of the nuclear bile acid receptor FXR. Mol. Cell 11, 1079–1092 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Staudinger, J. L. et al. The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc. Natl Acad. Sci. USA 98, 3369–3374 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Xie, W. et al. An essential role for nuclear receptors SXR/PXR in detoxification of cholestatic bile acids. Proc. Natl Acad. Sci. USA 98, 3375–3380 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Makishima, M. et al. Vitamin D receptor as an intestinal bile acid sensor. Science 296, 1313–1316 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Zollner, G., Marschall, H. U., Wagner, M. & Trauner, M. Role of nuclear receptors in the adaptive response to bile acids and cholestasis: pathogenetic and therapeutic considerations. Mol. Pharm. 3, 231–251 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Eloranta, J. J. & Kullak-Ublick, G. A. Coordinate transcriptional regulation of bile acid homeostasis and drug metabolism. Arch. Biochem. Biophys. 433, 397–412 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Pircher, P. C. et al. Farnesoid X receptor regulates bile acid-amino acid conjugation. J. Biol. Chem. 278, 27703–27711 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Moschetta, A., Bookout, A. L. & Mangelsdorf, D. J. Prevention of cholesterol gallstone disease by FXR agonists in a mouse model. Nature Med. 10, 1352–1358 (2004). An elegant study demonstrating that the activation of the FXR-signalling pathway prevents the formation of cholesterol gallstones.

    Article  CAS  PubMed  Google Scholar 

  33. Chiang, J. Y. Bile acid regulation of gene expression: roles of nuclear hormone receptors. Endocr. Rev. 23, 443–463 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Grober, J. et al. Identification of a bile acid-responsive element in the human ileal bile acid-binding protein gene. Involvement of the farnesoid X receptor/9-cis-retinoic acid receptor heterodimer. J. Biol. Chem. 274, 29749–29754 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Landrier, J. F., Eloranta, J. J., Vavricka, S. R. & Kullak-Ublick, G. A. The nuclear receptor for bile acids, FXR, transactivates human organic solute transporter-α and -β genes. Am. J. Physiol. Gastrointest. Liver Physiol. 290, G476–G485 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Lee, H. et al. FXR regulates organic solute transporters alpha and beta in the adrenal gland, kidney, and intestine. J. Lipid Res. 47, 201–214 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Holt, J. A. et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 17, 1581–1591 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Inagaki, T. et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell. Metab. 2, 217–225 (2005). References 37 and 38 identify intestinal FGF15/FGF19 as a key regulator of bile-acid synthesis.

    Article  CAS  PubMed  Google Scholar 

  39. Bavner, A., Sanyal, S., Gustafsson, J. A. & Treuter, E. Transcriptional corepression by SHP: molecular mechanisms and physiological consequences. Trends Endocrinol. Metab. 16, 478–488 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Houten, S. M., Volle, D. H., Cummins, C. L., Mangelsdorf, D. J. & Auwerx, J. In vivo imaging of farnesoid X receptor activity reveals the ileum as the primary bile acid signaling tissue. Mol. Endocrinol. 21, 1312–1323 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Foord, S. M. et al. International Union of Pharmacology. XLVI. G protein-coupled receptor list. Pharmacol. Rev. 57, 279–288 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Watanabe, M. et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439, 484–489 (2006). A key paper identifying bile acids as endocrine factors that induce energy expenditure by promoting intracellular thyroid hormone activation through the activation of the TGR5-signalling pathway.

    Article  CAS  PubMed  Google Scholar 

  43. Vassileva, G. et al. Targeted deletion of Gpbar1 protects mice from cholesterol gallstone formation. Biochem. J. 398, 423–430 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Keitel, V. et al. The G-protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells. Hepatology 45, 695–704 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Katsuma, S., Hirasawa, A. & Tsujimoto, G. Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem. Biophys. Res. Commun. 329, 386–390 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Maruyama, T. et al. Targeted disruption of G protein-coupled bile acid receptor 1 (Gpbar1/M-Bar) in mice. J. Endocrinol. 191, 197–205 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Yang, J. I. et al. Bile acid-induced TGR5-dependent c-Jun-N terminal kinase activation leads to enhanced caspase 8 activation in hepatocytes. Biochem. Biophys. Res. Commun. 361, 156–161 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Yasuda, H. et al. Involvement of membrane-type bile acid receptor M-BAR/TGR5 in bile acid-induced activation of epidermal growth factor receptor and mitogen-activated protein kinases in gastric carcinoma cells. Biochem. Biophys. Res. Commun. 354, 154–159 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Lu, T. T. et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol. Cell 6, 507–515 (2000).

    Article  CAS  PubMed  Google Scholar 

  50. Goodwin, B. et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol. Cell 6, 517–526 (2000). References49 and 50 illustrate the molecular basis for feedback regulation of bile-acid biosynthesis.

    Article  CAS  PubMed  Google Scholar 

  51. Brendel, C., Schoonjans, K., Botrugno, O. A., Treuter, E. & Auwerx, J. The small heterodimer partner (SHP) interacts with the Liver X receptor α (LXRα) and represses its transcriptional activity. Mol. Endocrinol. 16, 2065–2076 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Mataki, C. et al. Compromised intestinal lipid absorption in mice with a liver-specific deficiency of liver receptor homolog 1. Mol. Cell Biol. 27, 8330–8339 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lee, Y. K. et al. Liver receptor homolog-1 regulates bile acid homeostasis but is not essential for feedback regulation of bile acid synthesis. Mol. Endocrinol. 22, 1345–1356 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Stroup, D., Crestani, M. & Chiang, J. Y. Identification of a bile acid response element in the cholesterol 7 α-hydroxylase gene CYP7A. Am. J. Physiol. 273, G508–G517 (1997).

    CAS  PubMed  Google Scholar 

  55. De Fabiani, E. et al. The negative effects of bile acids and tumor necrosis factor-α on the transcription of cholesterol 7α-hydroxylase gene (CYP7A1) converge to hepatic nuclear factor-4: a novel mechanism of feedback regulation of bile acid synthesis mediated by nuclear receptors. J. Biol. Chem. 276, 30708–30716 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Zhang, M. & Chiang, J. Y. Transcriptional regulation of the human sterol 12α-hydroxylase gene (CYP8B1): roles of hepatocyte nuclear factor 4α in mediating bile acid repression. J. Biol. Chem. 276, 41690–41699 (2001).

    Article  CAS  PubMed  Google Scholar 

  57. Inoue, Y. et al. Regulation of bile acid biosynthesis by hepatocyte nuclear factor 4α. J. Lipid Res. 47, 215–227 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Choi, M. et al. Identification of a hormonal basis for gallbladder filling. Nature Med. 12, 1253–1255 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Ito, S. et al. Impaired negative feedback suppression of bile acid synthesis in mice lacking βKlotho. J. Clin. Invest. 115, 2202–2208 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Goetz, R. et al. Molecular insights into the klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members. Mol. Cell Biol. 27, 3417–3428 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wu, X. et al. Co-receptor requirements for fibroblast growth factor-19 signaling. J. Biol. Chem. 282, 29069–29072 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Lin, B. C., Wang, M., Blackmore, C. & Desnoyers, L. R. Liver-specific activities of FGF19 require Klotho β. J. Biol. Chem. 282, 27277–27284 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Goodwin, B. et al. Differential regulation of rat and human CYP7A1 by the nuclear oxysterol receptor liver X receptor-α. Mol. Endocrinol. 17, 386–394 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Schoenfield, L. J. & Lachin, J. M. Chenodiol (chenodeoxycholic acid) for dissolution of gallstones: the National Cooperative Gallstone Study. A controlled trial of efficacy and safety. Ann. Intern. Med. 95, 257–282 (1981).

    Article  CAS  PubMed  Google Scholar 

  65. Leiss, O. & von Bergmann, K. Different effects of chenodeoxycholic acid and ursodeoxycholic acid on serum lipoprotein concentrations in patients with radiolucent gallstones. Scand. J. Gastroenterol. 17, 587–592 (1982).

    Article  CAS  PubMed  Google Scholar 

  66. Francis, G. A., Fayard, E., Picard, F. & Auwerx, J. Nuclear receptors and the control of metabolism. Annu. Rev. Physiol. 65, 261–311 (2003).

    Article  CAS  PubMed  Google Scholar 

  67. Grundy, S. M., Ahrens, E. H. Jr & Salen, G. Interruption of the enterohepatic circulation of bile acids in man: comparative effects of cholestyramine and ileal exclusion on cholesterol metabolism. J. Lab. Clin. Med. 78, 94–121 (1971).

    CAS  PubMed  Google Scholar 

  68. Nestel, P. J. & Grundy, S. M. Changes in plasma triglyceride metabolism during withdrawal of bile. Metabolism 25, 1259–1268 (1976).

    Article  CAS  PubMed  Google Scholar 

  69. Angelin, B., Einarsson, K., Hellstrom, K. & Leijd, B. Effects of cholestyramine and chenodeoxycholic acid on the metabolism of endogenous triglyceride in hyperlipoproteinemia. J. Lipid Res. 19, 1017–1024 (1978).

    Article  CAS  PubMed  Google Scholar 

  70. Kobayashi, M. et al. Prevention and treatment of obesity, insulin resistance, and diabetes by bile acid-binding resin. Diabetes 56, 239–247 (2007). Reports the key observation that treatment with bile-acid-binding resin reduces obesity and insulin resistance in mice.

    Article  CAS  PubMed  Google Scholar 

  71. Miller, N. E. & Nestel, P. J. Triglyceride-lowering effect of chenodeoxycholic acid in patients with endogenous hypertriglyceridaemia. Lancet 2, 929–931 (1974).

    Article  CAS  PubMed  Google Scholar 

  72. Kast, H. R. et al. Farnesoid X-activated receptor induces apolipoprotein C-II transcription: a molecular mechanism linking plasma triglyceride levels to bile acids. Mol. Endocrinol. 15, 1720–1728 (2001).

    Article  CAS  PubMed  Google Scholar 

  73. Kanaya, E., Shiraki, T. & Jingami, H. The nuclear bile acid receptor FXR is activated by PGC-1α in a ligand-dependent manner. Biochem. J. 382, 913–921 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Stayrook, K. R. et al. Regulation of carbohydrate metabolism by the farnesoid X receptor. Endocrinology 146, 984–991 (2005).

    Article  CAS  PubMed  Google Scholar 

  75. Watanabe, M. et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J. Clin. Invest. 113, 1408–1418 (2004). This study describes the molecular mechanism underlying the triglyceride-lowering action of bile acids.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhang, Y. et al. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc. Natl Acad. Sci. USA 103, 1006–1011 (2006). An interesting study demonstrating that the FXR-signalling pathway controls glucose and lipid homeostasis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Bilz, S. et al. Activation of the farnesoid X receptor improves lipid metabolism in combined hyperlipidemic hamsters. Am. J. Physiol. Endocrinol. Metab. 290, E716–E722 (2006).

    Article  CAS  PubMed  Google Scholar 

  78. Horton, J. D., Goldstein, J. L. & Brown, M. S. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109, 1125–1131 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Shimomura, I. et al. Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proc. Natl Acad. Sci. USA 96, 13656–13661 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Foretz, M., Guichard, C., Ferré, P. & Foufelle, F. Sterol regulatory element binding protein 1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes. Proc. Natl Acad. Sci. USA 96, 12737–12742 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Schultz, J. R. et al. Role of LXRs in control of lipogenesis. Genes Dev. 14, 2831–2838 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Repa, J. J. et al. Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRα and LXRβ. Genes Dev. 14, 2819–2830 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Nishigori, H. et al. Mutations in the small heterodimer partner gene are associated with mild obesity in Japanese subjects. Proc. Natl Acad. Sci. USA 98, 575–580 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hung., C. C. et al. Contribution of variants in the small heterodimer partner gene to birthweight, adiposity, and insulin levels: mutational analysis and association studies in multiple populations. Diabetes 52, 1288–1291 (2003).

    Article  CAS  PubMed  Google Scholar 

  85. Mitchell, S. M. et al. Genetic variation in the small heterodimer partner gene and young-onset type 2 diabetes, obesity, and birth weight in U.K. subjects. Diabetes 52, 1276–1279 (2003).

    Article  CAS  PubMed  Google Scholar 

  86. Wang, L. et al. Redundant pathways for negative feedback regulation of bile acid production. Dev. Cell 2, 721–731 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Kerr, T. A. et al. Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev. Cell 2, 713–720 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Sinal, C. J. et al. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102, 731–744 (2000). Demonstrates that FXR is a crucial regulator of bile-acid homeostasis.

    Article  CAS  PubMed  Google Scholar 

  89. Zhang, Y. & Edwards, P. A. FXR signaling in metabolic disease. FEBS Lett. 582, 10–18 (2008).

    Article  CAS  PubMed  Google Scholar 

  90. Ikemoto, S. et al. Cholate inhibits high-fat diet-induced hyperglycemia and obesity with acyl-CoA synthetase mRNA decrease. Am. J. Physiol. 273, E37–E45 (1997).

    Article  CAS  PubMed  Google Scholar 

  91. Sato, H. et al. Anti-hyperglycemic activity of a TGR5 agonist isolated from Olea Europea. Biochem. Biophys. Res. Commun. 362, 793–798 (2007).

    Article  CAS  PubMed  Google Scholar 

  92. Tomlinson, E. et al. Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology 143, 1741–1747 (2002).

    Article  CAS  PubMed  Google Scholar 

  93. Wang, L. et al. The orphan nuclear receptor SHP regulates PGC-1α expression and energy production in brown adipocytes. Cell. Metab. 2, 227–238 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Garg, A. & Grundy, S. M. Cholestyramine therapy for dyslipidemia in non-insulin-dependent diabetes mellitus. A short-term, double-blind, crossover trial. Ann. Intern. Med. 121, 416–422 (1994).

    Article  CAS  PubMed  Google Scholar 

  95. Duran-Sandoval, D. et al. The farnesoid X receptor modulates hepatic carbohydrate metabolism during the fasting-refeeding transition. J. Biol. Chem. 280, 29971–29979 (2005).

    Article  CAS  PubMed  Google Scholar 

  96. De Fabiani, E. et al. Coordinated control of cholesterol catabolism to bile acids and of gluconeogenesis via a novel mechanism of transcription regulation linked to the fasted-to-fed cycle. J. Biol. Chem. 278, 39124–39132 (2003).

    Article  CAS  PubMed  Google Scholar 

  97. Yamagata, K. et al. Bile acids regulate gluconeogenic gene expression via small heterodimer partner-mediated repression of hepatocyte nuclear factor 4 and Foxo1. J. Biol. Chem. 279, 23158–23165 (2004).

    Article  CAS  PubMed  Google Scholar 

  98. Ma, K., Saha, P. K., Chan, L. & Moore, D. D. Farnesoid X receptor is essential for normal glucose homeostasis. J. Clin. Invest. 116, 1102–1109 (2006). An interesting study demonstrating that the FXR signalling pathway controls glucose homeostasis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Koo, S. H. et al. PGC-1 promotes insulin resistance in liver through PPAR-α-dependent induction of TRB-3. Nature Med. 10, 530–534 (2004).

    Article  CAS  PubMed  Google Scholar 

  100. Cariou, B. et al. Transient impairment of the adaptive response to fasting in FXR-deficient mice. FEBS Lett. 579, 4076–4080 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. Cariou, B. et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J. Biol. Chem. 281, 11039–11049 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Han, S. I. et al. Bile acids enhance the activity of the insulin receptor and glycogen synthase in primary rodent hepatocytes. Hepatology 39, 456–463 (2004).

    Article  CAS  PubMed  Google Scholar 

  103. Fang, Y. et al. Bile acids induce mitochondrial ROS, which promote activation of receptor tyrosine kinases and signaling pathways in rat hepatocytes. Hepatology 40, 961–971 (2004).

    Article  CAS  PubMed  Google Scholar 

  104. Dent, P. et al. Conjugated bile acids promote ERK1/2 and AKT activation via a pertussis toxin-sensitive mechanism in murine and human hepatocytes. Hepatology 42, 1291–1299 (2005).

    Article  CAS  PubMed  Google Scholar 

  105. Fang, Y. et al. Conjugated bile acids regulate hepatocyte glycogen synthase activity in vitro and in vivo via Gαi signaling. Mol. Pharmacol. 71, 1122–1128 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. Auwerx, J. Improving metabolism by increasing energy expenditure. Nature Med. 12, 44–45; discussion 45 (2006).

    Article  CAS  PubMed  Google Scholar 

  107. Ozcan, U. et al. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 313, 1137–1140 (2006). An interesting study demonstrating that the bile acid tauroursodeoxycholic acid restores glucose homeo-stasis by reducing endoplasmic reticulum stress.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Fiorucci, S., Rizzo, G., Donini, A., Distrutti, E. & Santucci, L. Targeting farnesoid X receptor for liver and metabolic disorders. Trends Mol. Med. 13, 298–309 (2007).

    Article  CAS  PubMed  Google Scholar 

  109. Inagaki, T. et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc. Natl Acad. Sci. USA 103, 3920–3925 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Jiang, T. et al. Farnesoid X receptor modulates renal lipid metabolism, fibrosis, and diabetic nephropathy. Diabetes 56, 2485–2493 (2007).

    Article  CAS  PubMed  Google Scholar 

  111. Maloney, P. R. et al. Identification of a chemical tool for the orphan nuclear receptor FXR. J. Med. Chem. 43, 2971–2974 (2000).

    Article  CAS  PubMed  Google Scholar 

  112. Liu, Y. et al. Hepatoprotection by the farnesoid X receptor agonist GW4064 in rat models of intra- and extrahepatic cholestasis. J. Clin. Invest. 112, 1678–1687 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Harnish, D. C. et al. A synthetic farnesoid X receptor (FXR) agonist protects against diet-induced dyslipidemia. J. Clin. Lipidol. 1, 362 (2007).

    Google Scholar 

  114. Pellicciari, R., Costantino, G. & Fiorucci, S. Farnesoid X receptor: from structure to potential clinical applications. J. Med. Chem. 48, 5383–5403 (2005).

    Article  CAS  PubMed  Google Scholar 

  115. Cui, J. et al. Guggulsterone is a farnesoid X receptor antagonist in coactivator association assays but acts to enhance transcription of bile salt export pump. J. Biol. Chem. 278, 10214–10220 (2003).

    Article  CAS  PubMed  Google Scholar 

  116. Dussault, I. et al. Identification of gene-selective modulators of the bile acid receptor FXR. J. Biol. Chem. 278, 7027–7033 (2003).

    Article  CAS  PubMed  Google Scholar 

  117. Urizar, N. L. et al. A natural product that lowers cholesterol as an antagonist ligand for FXR. Science 296, 1703–1706 (2002). This study identifies guggulsterone as an FXR antagonist and relays this property to its cholesterol-lowering effects.

    Article  CAS  PubMed  Google Scholar 

  118. Wu, J. et al. The hypolipidemic natural product guggulsterone acts as an antagonist of the bile acid receptor. Mol. Endocrinol. 16, 1590–1597 (2002).

    Article  CAS  PubMed  Google Scholar 

  119. Szapary, P. O. et al. Guggulipid for the treatment of hypercholesterolemia: a randomized controlled trial. JAMA 290, 765–772 (2003).

    Article  CAS  PubMed  Google Scholar 

  120. Meyer, U., Costantino, G., Macchiarulo, A. & Pellicciari, R. Is antagonism of E/Z-guggulsterone at the farnesoid X receptor mediated by a noncanonical binding site? A molecular modeling study. J. Med. Chem. 48, 6948–6955 (2005).

    Article  CAS  PubMed  Google Scholar 

  121. Sato, H. et al. Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structure–activity relationships, and molecular modeling studies. J. Med. Chem. 51, 1831–1841 (2008). The first description of the structure–activity relationship of bile acids as TGR5 ligands.

    Article  CAS  PubMed  Google Scholar 

  122. Pellicciari, R. et al. Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23- and 6,23-alkyl-substituted bile acid derivatives as selective modulators for the G-protein coupled receptor TGR5. J. Med. Chem. 50, 4265–4268 (2007).

    Article  CAS  PubMed  Google Scholar 

  123. Fumio, I. et al., Receptor antagonists. WO2004067008 (2004).

  124. Tsui, P. & Michalovich, D. G protein coupled receptor AXOR 109. GB2371546A (2001).

  125. Yamakawa, T., Takano, T., Utsunomiya, H., Kadonosono, K. & Okamura, A. Effect of colestimide therapy for glycemic control in type 2 diabetes mellitus with hypercholesterolemia. Endocr. J. 54, 53–58 (2007).

    Article  CAS  PubMed  Google Scholar 

  126. Sakamoto, S. et al. Glucuronidation converting methyl 1-(3,4-dimethoxyphenyl)-3-(3-ethylvaleryl)-4-hydroxy-6,7,8-trimethoxy-2-na phthoate (S-8921) to a potent apical sodium-dependent bile acid transporter inhibitor, resulting in a hypocholesterolemic action. J. Pharmacol. Exp. Ther. 322, 610–618 (2007).

    Article  CAS  PubMed  Google Scholar 

  127. Li, H. et al. Inhibition of ileal bile acid transport lowers plasma cholesterol levels by inactivating hepatic farnesoid X receptor and stimulating cholesterol 7 α-hydroxylase. Metabolism 53, 927–932 (2004).

    Article  CAS  PubMed  Google Scholar 

  128. West, K. L., Zern, T. L., Butteiger, D. N., Keller, B. T. & Fernandez, M. L. SC-435, an ileal apical sodium co-dependent bile acid transporter (ASBT) inhibitor lowers plasma cholesterol and reduces atherosclerosis in guinea pigs. Atherosclerosis 171, 201–210 (2003).

    Article  CAS  PubMed  Google Scholar 

  129. 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 

  130. Martin, F. P. et al. A top-down systems biology view of microbiome–mammalian metabolic interactions in a mouse model. Mol. Syst. Biol. 3, 112 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Li, M. et al. Symbiotic gut microbes modulate human metabolic phenotypes. Proc. Natl Acad. Sci. USA 105, 2117–2122 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Libert, S. & Pletcher, S. D. Modulation of longevity by environmental sensing. Cell 131, 1231–1234 (2007).

    Article  CAS  PubMed  Google Scholar 

  133. Holst, J. J. The physiology of glucagon-like peptide 1. Physiol. Rev. 87, 1409–1439 (2007).

    Article  CAS  PubMed  Google Scholar 

  134. Huang, W. et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 312, 233–236 (2006).

    Article  CAS  PubMed  Google Scholar 

  135. Motola, D. L. et al. Identification of ligands for DAF-12 that govern dauer formation and reproduction in C. elegans. Cell 124, 1209–1223 (2006).

    Article  CAS  PubMed  Google Scholar 

  136. Gerisch, B. et al. A bile acid-like steroid modulates Caenorhabditis elegans lifespan through nuclear receptor signaling. Proc. Natl Acad. Sci. USA 104, 5014–5019 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang, D. Q., Tazuma, S., Cohen, D. E. & Carey, M. C. Feeding natural hydrophilic bile acids inhibits intestinal cholesterol absorption: studies in the gallstone-susceptible mouse. Am. J. Physiol. Gastrointest. Liver Physiol. 285, G494–G502 (2003).

    Article  CAS  PubMed  Google Scholar 

  138. Raufman, J. P., Zimniak, P. & Bartoszko-Malik, A. Lithocholyltaurine interacts with cholinergic receptors on dispersed chief cells from guinea pig stomach. Am. J. Physiol. 274, G997–G1004 (1998).

    CAS  PubMed  Google Scholar 

  139. Wess, J., Eglen, R. M. & Gautam, D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nature Rev. Drug Discov. 6, 721–733 (2007).

    Article  CAS  Google Scholar 

  140. Gautam, D. et al. A critical role for beta cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell. Metab. 3, 449–461 (2006).

    Article  CAS  PubMed  Google Scholar 

  141. Le, Y., Murphy, P. M. & Wang, J. M. Formyl-peptide receptors revisited. Trends Immunol. 23, 541–548 (2002).

    Article  CAS  PubMed  Google Scholar 

  142. Chen, X. et al. Characterization of chenodeoxycholic acid as an endogenous antagonist of the G-coupled formyl peptide receptors. Inflamm. Res. 49, 744–755 (2000).

    Article  CAS  PubMed  Google Scholar 

  143. Podevin, P. et al. Effect of cholestasis and bile acids on interferon-induced 2′,5′-adenylate synthetase and NK cell activities. Gastroenterology 108, 1192–1198 (1995).

    Article  CAS  PubMed  Google Scholar 

  144. Ferrari, C., Macchiarulo, A., Costantino, G. & Pellicciari, R. Pharmacophore model for bile acids recognition by the FPR receptor. J. Comput. Aided Mol. Des. 20, 295–303 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Qiao, L. et al. Bile acid regulation of C/EBPβ, CREB, and c-Jun function, via the extracellular signal-regulated kinase and c-Jun NH2-terminal kinase pathways, modulates the apoptotic response of hepatocytes. Mol. Cell Biol. 23, 3052–3066 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Kim, K. M. et al. Bile acid-mediated induction of cyclooxygenase-2 and Mcl-1 in hepatic stellate cells. Biochem. Biophys. Res. Commun. 342, 1108–1113 (2006).

    Article  CAS  PubMed  Google Scholar 

  147. Qiao, D., Stratagouleas, E. D. & Martinez, J. D. Activation and role of mitogen-activated protein kinases in deoxycholic acid-induced apoptosis. Carcinogenesis 22, 35–41 (2001).

    Article  CAS  PubMed  Google Scholar 

  148. Gupta, S., Stravitz, R. T., Dent, P. & Hylemon, P. B. Down-regulation of cholesterol 7α-hydroxylase (CYP7A1) gene expression by bile acids in primary rat hepatocytes is mediated by the c-Jun N-terminal kinase pathway. J. Biol. Chem. 276, 15816–15822 (2001).

    Article  CAS  PubMed  Google Scholar 

  149. Stamp, D. H. Three hypotheses linking bile to carcinogenesis in the gastrointestinal tract: certain bile salts have properties that may be used to complement chemotherapy. Med. Hypotheses 59, 398–405 (2002).

    Article  CAS  PubMed  Google Scholar 

  150. Pandak, W. M. et al. Expression of sterol 12α-hydroxylase alters bile acid pool composition in primary rat hepatocytes and in vivo. Gastroenterology 120, 1801–1809 (2001).

    Article  CAS  PubMed  Google Scholar 

  151. Li-Hawkins, J. et al. Cholic acid mediates negative feedback regulation of bile acid synthesis in mice. J. Clin. Invest. 110, 1191–1200 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Falany, C. N. et al. Molecular cloning and expression of rat liver bile acid CoA ligase. J. Lipid Res. 43, 2062–2071 (2002).

    Article  CAS  PubMed  Google Scholar 

  153. Shonsey, E. M. et al. Bile acid coenzyme A: amino acid N-acyltransferase in the amino acid conjugation of bile acids. Methods Enzymol. 400, 374–394 (2005).

    Article  CAS  PubMed  Google Scholar 

  154. He, D., Barnes, S. & Falany, C. N. Rat liver bile acid CoA:amino acid N-acyltransferase: expression, characterization, and peroxisomal localization. J. Lipid Res. 44, 2242–2249 (2003).

    Article  CAS  PubMed  Google Scholar 

  155. Meier, P. J. & Stieger, B. Bile salt transporters. Annu. Rev. Physiol. 64, 635–661 (2002).

    Article  CAS  PubMed  Google Scholar 

  156. Trauner, M. & Boyer, J. L. Bile salt transporters: molecular characterization, function, and regulation. Physiol. Rev. 83, 633–671 (2003).

    Article  CAS  PubMed  Google Scholar 

  157. Rao, A. et al. The organic solute transporter α-β, Ostα-Ostβ, is essential for intestinal bile acid transport and homeostasis. Proc. Natl Acad. Sci. USA 105, 3891–3896 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Modica, S. & Moschetta, A. Nuclear bile acid receptor FXR as pharmacological target: are we there yet? FEBS Lett. 580, 5492–5499 (2006).

    Article  CAS  PubMed  Google Scholar 

  159. Pellicciari, R. et al. 6α-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. J. Med. Chem. 45, 3569–3572 (2002).

    Article  CAS  PubMed  Google Scholar 

  160. Cai, S. Y., Xiong, L., Wray, C. G., Ballatori, N. & Boyer, J. L. The farnesoid X receptor FXRα/NR1H4 acquired ligand specificity for bile salts late in vertebrate evolution. Am. J. Physiol. Regul. Integr. Comp. Physiol. 293, R1400–R1409 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support by grants from the Centre National de la Recherche Scientifique, the Institut National de la Santé et de la Recherche Médicale, the Université Louis Pasteur, the Hôpital Universitaire de Strasbourg, the National Institutes of Health (DK59820 and DK067320), L'Agence Nationale pour la Recherche (ANR 07-PHYSIO-003-01), La Fondation pour la Recherche Médicale, Ligue contre le Cancer, Intercept Pharmaceuticals (New York, USA), the Ecole Polytechnique Federale de Lausanne and the European Union (LSHM-CT-2004-512,013), and by fellowships from Société Française de Nutrition and L'Association pour la Recherche sur le Cancer (to C.T.). We apologize to those authors whose original work could not be quoted owing to space restrictions.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Johan Auwerx or Kristina Schoonjans.

Ethics declarations

Competing interests

J.A. and R.P. are consultants for Intercept Pharmaceuticals, and M.P. is CEO of Intercept Pharmaceuticals, a company that is developing FXR-α and TGR5 ligands.

Related links

Related links

FURTHER INFORMATION

ClinicalTrials.gov

Glossary

Bile canaliculus

One of the intercellular channels that occur between liver cells that form the first portion of the bile system. The different components of bile, synthesized and secreted by hepatocytes, are collected in these bile canaliculi, which merge and ultimately form bile ductules.

Atherosclerosis

A chronic inflammatory response in the walls of arteries, in large part due to the infiltration of macrophages, which accumulate cholesterol from low-density lipoproteins. Without the adequate removal of this cholesterol by high-density lipoproteins, these macrophages become foam cells.

Cerebrotendinous xanthomatosis

An inherited disorder that is associated with elevated circulating cholesterol levels and deposition of cholestanol in the brain and other tissues. This disorder is characterized by progressive cerebellar ataxia beginning after puberty, by juvenile cataracts, and by tendinous or tuberous xanthomas.

Syntenic

Describes a preserved co-localization of genes on chromosomes between different species.

Brown adipose tissue

Brown adipose tissue (BAT) is one of the two types of adipose tissue, the other being white adipose tissue. BAT is present in human newborns or small mammals. Its primary purpose is to generate body heat, owing to a high density of mitochondria.

Lithogenic diet

An experimental diet to induce cholelithiasis or cholesterol gallstones in the gall bladder.

Kupffer cells

Kupffer cells are specialized macrophages located in the liver that are part of the reticuloendothelial system.

Incretins

Incretins are gastrointestinal hormones that cause an increase in the amount of insulin released from the pancreatic β-cells after a meal. Gastric inhibitory peptide (GIP) — also known as glucose-dependent insulinotropic peptide — and glucagon-like peptide 1 (GLP1) are the two main incretins secreted by the enteroendocrine K and L cells, respectively.

Hypertriglyceridaemia

Denotes high levels of triglycerides in the blood.

Type 2 iodothyronine deiodinase

This enzyme activates thyroid hormone by converting the prohormone thyroxine (T4) by outer ring deiodination to the bioactive 3,3′,5-triiodothyronine (T3).

Diabesity

An association of obesity and type 2 diabetes.

Endoplasmic reticulum stress

Cellular disturbances causing an accumulation of unfolded proteins in the endoplasmic reticulum, which results in activation of the unfolded protein response.

Non-alcoholic steato-hepatitis

Non-alcoholic steatohepatitis (NASH) is a common and often silent liver disease, which has become one of the most prevalent alcohol-independent causes of liver cirrhosis in Western countries. Similar to non-alcoholic fatty liver disease (NAFLD), NASH is characterized by the accumulation of hepatic fat droplets, but distinguishes itself from NAFLD by the presence of a significant inflammatory component.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Thomas, C., Pellicciari, R., Pruzanski, M. et al. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov 7, 678–693 (2008). https://doi.org/10.1038/nrd2619

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2619

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