Skip to main content
Top
Published in: Current Diabetes Reports 11/2018

01-11-2018 | Pathogenesis of Type 2 Diabetes and Insulin Resistance (M-E Patti, Section Editor)

Rethinking Bile Acid Metabolism and Signaling for Type 2 Diabetes Treatment

Authors: Karolina E. Zaborska, Bethany P. Cummings

Published in: Current Diabetes Reports | Issue 11/2018

Login to get access

Abstract

Purpose of Review

Herein, we review the role of FXR and TGR5 in the regulation of hepatic bile acid metabolism, with a focus on how our understanding of bile acid metabolic regulation by these receptors has evolved in recent years and how this improved understanding may facilitate targeting bile acids for type 2 diabetes treatment.

Recent Findings

Bile acid profile is a key regulator of metabolic homeostasis. Inhibition of expression of the enzyme that is required for cholic acid synthesis and thus determines bile acid profile, Cyp8b1, may be an effective target for type 2 diabetes treatment. FXR and, more recently, TGR5 have been shown to regulate bile acid metabolism and Cyp8b1 expression and, therefore, may provide a mechanism with which to target bile acid profile for type 2 diabetes treatment.

Summary

Inhibition of Cyp8b1 expression is a promising therapeutic modality for type 2 diabetes; however, further work is needed to fully understand the pathways regulating Cyp8b1 expression.
Literature
4.
go back to reference Heuman DM. Quantitative estimation of the hydrophilic-hydrophobic balance of mixed bile salt solutions. J Lipid Res. 1989;30(5):719–30.PubMed Heuman DM. Quantitative estimation of the hydrophilic-hydrophobic balance of mixed bile salt solutions. J Lipid Res. 1989;30(5):719–30.PubMed
7.
23.
go back to reference Hagey LR, Crombie DL, Espinosa E, Carey MC, Igimi H, Hofmann AF. Ursodeoxycholic acid in the Ursidae: biliary bile acids of bears, pandas, and related carnivores. J Lipid Res. 1993;34(11):1911–7.PubMed Hagey LR, Crombie DL, Espinosa E, Carey MC, Igimi H, Hofmann AF. Ursodeoxycholic acid in the Ursidae: biliary bile acids of bears, pandas, and related carnivores. J Lipid Res. 1993;34(11):1911–7.PubMed
24.
go back to reference Bachrach WH, Hofmann AF. Ursodeoxycholic acid in the treatment of cholesterol cholelithiasis. part I. Dig Dis Sci. 1982;27(8):737–61.CrossRefPubMed Bachrach WH, Hofmann AF. Ursodeoxycholic acid in the treatment of cholesterol cholelithiasis. part I. Dig Dis Sci. 1982;27(8):737–61.CrossRefPubMed
25.
go back to reference Fedorowski T, Salen G, Tint GS, Mosbach E. Transformation of chenodeoxycholic acid and ursodeoxycholic acid by human intestinal bacteria. Gastroenterology. 1979;77(5):1068–73.PubMed Fedorowski T, Salen G, Tint GS, Mosbach E. Transformation of chenodeoxycholic acid and ursodeoxycholic acid by human intestinal bacteria. Gastroenterology. 1979;77(5):1068–73.PubMed
28.
go back to reference Norlin M, Wikvall K. Enzymes in the conversion of cholesterol into bile acids. Curr Mol Med. 2007;7(2):199–218.CrossRefPubMed Norlin M, Wikvall K. Enzymes in the conversion of cholesterol into bile acids. Curr Mol Med. 2007;7(2):199–218.CrossRefPubMed
29.
go back to reference Wu Z, Martin KO, Javitt NB, Chiang JY. Structure and functions of human oxysterol 7alpha-hydroxylase cDNAs and gene CYP7B1. J Lipid Res. 1999;40(12):2195–203.PubMed Wu Z, Martin KO, Javitt NB, Chiang JY. Structure and functions of human oxysterol 7alpha-hydroxylase cDNAs and gene CYP7B1. J Lipid Res. 1999;40(12):2195–203.PubMed
30.
go back to reference Andersson S, Davis DL, Dahlback H, Jornvall H, Russell DW. Cloning, structure, and expression of the mitochondrial cytochrome P-450 sterol 26-hydroxylase, a bile acid biosynthetic enzyme. J Biol Chem. 1989;264(14):8222–9.PubMed Andersson S, Davis DL, Dahlback H, Jornvall H, Russell DW. Cloning, structure, and expression of the mitochondrial cytochrome P-450 sterol 26-hydroxylase, a bile acid biosynthetic enzyme. J Biol Chem. 1989;264(14):8222–9.PubMed
32.
go back to reference Reiss AB, Martin KO, Rojer DE, Iyer S, Grossi EA, Galloway AC, et al. Sterol 27-hydroxylase: expression in human arterial endothelium. J Lipid Res. 1997;38(6):1254–60.PubMed Reiss AB, Martin KO, Rojer DE, Iyer S, Grossi EA, Galloway AC, et al. Sterol 27-hydroxylase: expression in human arterial endothelium. J Lipid Res. 1997;38(6):1254–60.PubMed
33.
go back to reference Schwarz M, Lund EG, Lathe R, Bjorkhem I, Russell DW. Identification and characterization of a mouse oxysterol 7alpha-hydroxylase cDNA. J Biol Chem. 1997;272(38):23995–4001..CrossRefPubMed Schwarz M, Lund EG, Lathe R, Bjorkhem I, Russell DW. Identification and characterization of a mouse oxysterol 7alpha-hydroxylase cDNA. J Biol Chem. 1997;272(38):23995–4001..CrossRefPubMed
34.
go back to reference Shanahan CM, Carpenter KL, Cary NR. A potential role for sterol 27-hydroxylase in atherogenesis. Atherosclerosis. 2001;154(2):269–76.CrossRefPubMed Shanahan CM, Carpenter KL, Cary NR. A potential role for sterol 27-hydroxylase in atherogenesis. Atherosclerosis. 2001;154(2):269–76.CrossRefPubMed
35.
go back to reference Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T, et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell. 1995;81(5):687–93.CrossRefPubMed Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T, et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell. 1995;81(5):687–93.CrossRefPubMed
37.
go back to reference De Gottardi A, Touri F, Maurer CA, Perez A, Maurhofer O, Ventre G, et al. The bile acid nuclear receptor FXR and the bile acid binding protein IBABP are differently expressed in colon cancer. Dig Dis Sci. 2004;49(6):982–9.CrossRefPubMed De Gottardi A, Touri F, Maurer CA, Perez A, Maurhofer O, Ventre G, et al. The bile acid nuclear receptor FXR and the bile acid binding protein IBABP are differently expressed in colon cancer. Dig Dis Sci. 2004;49(6):982–9.CrossRefPubMed
39.
go back to reference Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, et al. Identification of a nuclear receptor for bile acids. Science. 1999;284(5418):1362–5.CrossRefPubMed Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, et al. Identification of a nuclear receptor for bile acids. Science. 1999;284(5418):1362–5.CrossRefPubMed
40.
go back to reference Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell. 1999;3(5):543–53.CrossRefPubMed Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell. 1999;3(5):543–53.CrossRefPubMed
47.
go back to reference Fiorucci S, Antonelli E, Rizzo G, Renga B, Mencarelli A, Riccardi L, et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology. 2004;127(5):1497–512.CrossRefPubMed Fiorucci S, Antonelli E, Rizzo G, Renga B, Mencarelli A, Riccardi L, et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology. 2004;127(5):1497–512.CrossRefPubMed
57.
go back to reference Becker-Andre M, Andre E, DeLamarter JF. Identification of nuclear receptor mRNAs by RT-PCR amplification of conserved zinc-finger motif sequences. Biochem Biophys Res Commun. 1993;194(3):1371–9.CrossRefPubMed Becker-Andre M, Andre E, DeLamarter JF. Identification of nuclear receptor mRNAs by RT-PCR amplification of conserved zinc-finger motif sequences. Biochem Biophys Res Commun. 1993;194(3):1371–9.CrossRefPubMed
58.
go back to reference Chiang JY, Kimmel R, Weinberger C, Stroup D. Farnesoid X receptor responds to bile acids and represses cholesterol 7alpha-hydroxylase gene (CYP7A1) transcription. J Biol Chem. 2000;275(15):10918–24.CrossRefPubMed Chiang JY, Kimmel R, Weinberger C, Stroup D. Farnesoid X receptor responds to bile acids and represses cholesterol 7alpha-hydroxylase gene (CYP7A1) transcription. J Biol Chem. 2000;275(15):10918–24.CrossRefPubMed
59.
go back to reference Galarneau L, Pare JF, Allard D, Hamel D, Levesque L, Tugwood JD, et al. The alpha1-fetoprotein locus is activated by a nuclear receptor of the Drosophila FTZ-F1 family. Mol Cell Biol. 1996;16(7):3853–65.CrossRefPubMedPubMedCentral Galarneau L, Pare JF, Allard D, Hamel D, Levesque L, Tugwood JD, et al. The alpha1-fetoprotein locus is activated by a nuclear receptor of the Drosophila FTZ-F1 family. Mol Cell Biol. 1996;16(7):3853–65.CrossRefPubMedPubMedCentral
60.
go back to reference Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell. 2000;6(3):517–26.CrossRefPubMed Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell. 2000;6(3):517–26.CrossRefPubMed
61.
go back to reference Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell. 2000;6(3):507–15.CrossRefPubMed Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell. 2000;6(3):507–15.CrossRefPubMed
62.
go back to reference Nitta M, Ku S, Brown C, Okamoto AY, Shan B. CPF: an orphan nuclear receptor that regulates liver-specific expression of the human cholesterol 7alpha-hydroxylase gene. Proc Natl Acad Sci U S A. 1999;96(12):6660–5.CrossRefPubMedPubMedCentral Nitta M, Ku S, Brown C, Okamoto AY, Shan B. CPF: an orphan nuclear receptor that regulates liver-specific expression of the human cholesterol 7alpha-hydroxylase gene. Proc Natl Acad Sci U S A. 1999;96(12):6660–5.CrossRefPubMedPubMedCentral
63.
go back to reference Stroup D, Chiang JY. HNF4 and COUP-TFII interact to modulate transcription of the cholesterol 7alpha-hydroxylase gene (CYP7A1). J Lipid Res. 2000;41(1):1–11.PubMed Stroup D, Chiang JY. HNF4 and COUP-TFII interact to modulate transcription of the cholesterol 7alpha-hydroxylase gene (CYP7A1). J Lipid Res. 2000;41(1):1–11.PubMed
66.
go back to reference Yang Y, Zhang M, Eggertsen G, Chiang JY. On the mechanism of bile acid inhibition of rat sterol 12alpha-hydroxylase gene (CYP8B1) transcription: roles of alpha-fetoprotein transcription factor and hepatocyte nuclear factor 4alpha. Biochim Biophys Acta. 2002;1583(1):63–73.CrossRefPubMed Yang Y, Zhang M, Eggertsen G, Chiang JY. On the mechanism of bile acid inhibition of rat sterol 12alpha-hydroxylase gene (CYP8B1) transcription: roles of alpha-fetoprotein transcription factor and hepatocyte nuclear factor 4alpha. Biochim Biophys Acta. 2002;1583(1):63–73.CrossRefPubMed
71.
go back to reference Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, et al. Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell. 2002;2(6):713–20.CrossRefPubMedPubMedCentral Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, et al. Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell. 2002;2(6):713–20.CrossRefPubMedPubMedCentral
72.
go back to reference Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim CS, et al. Redundant pathways for negative feedback regulation of bile acid production. Dev Cell. 2002;2(6):721–31.CrossRefPubMed Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim CS, et al. Redundant pathways for negative feedback regulation of bile acid production. Dev Cell. 2002;2(6):721–31.CrossRefPubMed
73.
go back to reference • Xu Y, Li F, Zalzala M, Xu J, Gonzalez FJ, Adorini L, et al. Farnesoid X receptor activation increases reverse cholesterol transport by modulating bile acid composition and cholesterol absorption in mice. Hepatology. 2016;64(4):1072–85. https://doi.org/10.1002/hep.28712 This study provides important in vivo tissue-specific information on FXR regulation of hepatic bile acid metabolism. CrossRefPubMed • Xu Y, Li F, Zalzala M, Xu J, Gonzalez FJ, Adorini L, et al. Farnesoid X receptor activation increases reverse cholesterol transport by modulating bile acid composition and cholesterol absorption in mice. Hepatology. 2016;64(4):1072–85. https://​doi.​org/​10.​1002/​hep.​28712 This study provides important in vivo tissue-specific information on FXR regulation of hepatic bile acid metabolism. CrossRefPubMed
75.
82.
go back to reference Noshiro M, Nishimoto M, Okuda K. Rat liver cholesterol 7 alpha-hydroxylase. Pretranslational regulation for circadian rhythm. J Biol Chem. 1990;265(17):10036–41.PubMed Noshiro M, Nishimoto M, Okuda K. Rat liver cholesterol 7 alpha-hydroxylase. Pretranslational regulation for circadian rhythm. J Biol Chem. 1990;265(17):10036–41.PubMed
94.
go back to reference Salen G, Nicolau G, Shefer S, Mosbach EH. Hepatic cholesterol metabolism in patients with gallstones. Gastroenterology. 1975;69(3):676–84.PubMed Salen G, Nicolau G, Shefer S, Mosbach EH. Hepatic cholesterol metabolism in patients with gallstones. Gastroenterology. 1975;69(3):676–84.PubMed
95.
go back to reference Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun. 2002;298(5):714–9.CrossRefPubMed Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun. 2002;298(5):714–9.CrossRefPubMed
127.
130.
134.
go back to reference Myronovych A, Salazar-Gonzalez RM, Ryan KK, Miles L, Zhang W, Jha P, et al. The role of small heterodimer partner in nonalcoholic fatty liver disease improvement after sleeve gastrectomy in mice. Obesity (Silver Spring). 2014;22(11):2301–11. https://doi.org/10.1002/oby.20890.CrossRef Myronovych A, Salazar-Gonzalez RM, Ryan KK, Miles L, Zhang W, Jha P, et al. The role of small heterodimer partner in nonalcoholic fatty liver disease improvement after sleeve gastrectomy in mice. Obesity (Silver Spring). 2014;22(11):2301–11. https://​doi.​org/​10.​1002/​oby.​20890.CrossRef
Metadata
Title
Rethinking Bile Acid Metabolism and Signaling for Type 2 Diabetes Treatment
Authors
Karolina E. Zaborska
Bethany P. Cummings
Publication date
01-11-2018
Publisher
Springer US
Published in
Current Diabetes Reports / Issue 11/2018
Print ISSN: 1534-4827
Electronic ISSN: 1539-0829
DOI
https://doi.org/10.1007/s11892-018-1092-3

Other articles of this Issue 11/2018

Current Diabetes Reports 11/2018 Go to the issue

Pathogenesis of Type 1 Diabetes (A Pugliese and SJ Richardson, Section Editors)

The Role of Accessory Cells in Islet Homeostasis

Immunology, Transplantation, and Regenerative Medicine (L Piemonti and V Sordi, Section Editors)

Can We Re-Engineer the Endocrine Pancreas?

Health Care Delivery Systems and Implementation in Diabetes (ME McDonnell and AR Sadhu, Section Editors)

Building Toward a Population-Based Approach to Diabetes Screening and Prevention for US Adults

Other Forms of Diabetes and Its Complications (JJ Nolan and H Thabit, Section Editors)

Diabetes and HIV

Immunology, Transplantation, and Regenerative Medicine (L Piemonti and V Sordi, Section Editors)

Artificial Pancreas or Novel Beta-Cell Replacement Therapies: a Race for Optimal Glycemic Control?

Pathogenesis of Type 1 Diabetes (A Pugliese and SJ Richardson, Section Editors)

Pancreas Pathology During the Natural History of Type 1 Diabetes