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.

  • Letter
  • Published:

Central anorexigenic actions of bile acids are mediated by TGR5

Abstract

Bile acids (BAs) are signalling molecules that mediate various cellular responses in both physiological and pathological processes. Several studies report that BAs can be detected in the brain1, yet their physiological role in the central nervous system is still largely unknown. Here we show that postprandial BAs can reach the brain and activate a negative-feedback loop controlling satiety in response to physiological feeding via TGR5, a G-protein-coupled receptor activated by multiple conjugated and unconjugated BAs2 and an established regulator of peripheral metabolism3,4,5,6,7,8. Notably, peripheral or central administration of a BA mix or a TGR5-specific BA mimetic (INT-777) exerted an anorexigenic effect in wild-type mice, while whole-body, neuron-specific or agouti-related peptide neuronal TGR5 deletion caused a significant increase in food intake. Accordingly, orexigenic peptide expression and secretion were reduced after short-term TGR5 activation. In vitro studies demonstrated that activation of the Rho–ROCK–actin-remodelling pathway decreases orexigenic agouti-related peptide/neuropeptide Y (AgRP/NPY) release in a TGR5-dependent manner. Taken together, these data identify a signalling cascade by which BAs exert acute effects at the transition between fasting and feeding and prime the switch towards satiety, unveiling a previously unrecognized role of physiological feedback mediated by BAs in the central nervous system.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: BAs reach the hypothalamus during physiological feeding and suppress food intake through TGR5.
Fig. 2: TGR5 is expressed in neurons and its deletion increases food intake.
Fig. 3: Selective deletion of TGR5 in AgRP/NPY but not in POMC neurons abrogates satiety induced by BA derivatives.
Fig. 4: TGR5 regulates orexigenic neuropeptide secretion though transient activation of the Rho–ROCK signalling pathway.

Similar content being viewed by others

Data availability

All the datasets generated during the current study are available upon request to the corresponding author. Source data are provided with this paper.

References

  1. McMillin, M. & DeMorrow, S. Effects of bile acids on neurological function and disease. FASEB J. 30, 3658–3668 (2016).

    Article  CAS  Google Scholar 

  2. Kawamata, Y. et al. A G-protein-coupled receptor responsive to bile acids. J. Biol. Chem. 278, 9435–9440 (2003).

    Article  CAS  Google Scholar 

  3. Watanabe, M. et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439, 484–489 (2006).

    Article  CAS  Google Scholar 

  4. Thomas, C. et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10, 167–177 (2009).

    Article  CAS  Google Scholar 

  5. Pols, T. W. H. et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 14, 747–757 (2011).

    Article  CAS  Google Scholar 

  6. Perino, A. et al. TGR5 reduces macrophage migration through mTOR-induced C/EBPβ differential translation. J. Clin. Invest. 124, 5424–5436 (2014).

    Article  Google Scholar 

  7. Velazquez-Villegas, L. A. et al. TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat. Commun. 9, 245 (2018).

    Article  Google Scholar 

  8. Perino, A., Demagny, H., Velazquez-Villegas, L. A. & Schoonjans, K. Molecular physiology of bile acid signaling in health, disease and aging. Physiol. Rev. 101, 683–731 (2020).

    Article  Google Scholar 

  9. Angelin, B., Björkhem, I., Einarsson, K. & Ewerth, S. Hepatic uptake of bile acids in man. Fasting and postprandial concentrations of individual bile acids in portal venous and systemic blood serum. J. Clin. Invest. 70, 724–731 (1982).

    Article  CAS  Google Scholar 

  10. Higashi, T. et al. Unconjugated bile acids in rat brain: analytical method based on LC/ESI–MS/MS with chemical derivatization and estimation of their origin by comparison to serum levels. Steroids 125, 107–113 (2017).

    Article  CAS  Google Scholar 

  11. Mano, N. et al. Presence of protein-bound unconjugated bile acids in the cytoplasmic fraction of rat brain. J. Lipid Res. 45, 295–300 (2004).

    Article  CAS  Google Scholar 

  12. Zheng, X. et al. The brain metabolome of male rats across the lifespan. Sci. Rep. 6, 24125 (2016).

    Article  CAS  Google Scholar 

  13. Kiriyama, Y. & Nochi, H. The biosynthesis, signaling and neurological functions of bile acids. Biomolecules 9, 232 (2019).

    Article  CAS  Google Scholar 

  14. Lorbek, G., Lewinska, M. & Rozman, D. Cytochrome P450s in the synthesis of cholesterol and bile acids—from mouse models to human diseases. FEBS J. 279, 1516–1533 (2012).

    Article  CAS  Google Scholar 

  15. Yanguas-Casás, N., Barreda-Manso, M. A., Nieto-Sampedro, M. & Romero-Ramírez, L. TUDCA: an agonist of the bile acid receptor GPBAR1/TGR5 with anti-inflammatory effects in microglial cells. J. Cell. Physiol. 232, 2231–2245 (2017).

    Article  Google Scholar 

  16. Keitel, V. et al. The bile acid receptor TGR5 (Gpbar-1) acts as a neurosteroid receptor in brain. Glia 58, 1794–1805 (2010).

    Article  Google Scholar 

  17. Pellicciari, R. et al. Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity. J. Med. Chem. 52, 7958–7961 (2009).

    Article  CAS  Google Scholar 

  18. Waterson, M. J. & Horvath, T. L. Neuronal regulation of energy homeostasis: beyond the hypothalamus and feeding. Cell Metab. 22, 962–970 (2015).

    Article  CAS  Google Scholar 

  19. Schöne, C. & Burdakov, D. Glutamate and GABA as rapid effectors of hypothalamic ‘peptidergic’ neurons. Front. Behav. Neurosci. 6, 81 (2012).

    Article  Google Scholar 

  20. Meunier, F. A. & Gutiérrez, L. M. Captivating new roles of F-actin cortex in exocytosis and bulk endocytosis in neurosecretory cells. Trends Neurosci. 39, 605–613 (2016).

    Article  CAS  Google Scholar 

  21. Amano, M., Nakayama, M. & Kaibuchi, K. Rho-kinase/ROCK: a key regulator of the cytoskeleton and cell polarity. Cytoskeleton 67, 545–554 (2010).

    Article  CAS  Google Scholar 

  22. Sit, S.-T. & Manser, E. Rho GTPases and their role in organizing the actin cytoskeleton. J. Cell Sci. 124, 679–683 (2011).

    Article  CAS  Google Scholar 

  23. Symons, M. & Rusk, N. Control of vesicular trafficking by Rho GTPases. Curr. Biol. 13, R409–R418 (2003).

    Article  CAS  Google Scholar 

  24. Wolf, M. et al. ADF/cofilin controls synaptic actin dynamics and regulates synaptic vesicle mobilization and exocytosis. Cereb. Cortex 25, 2863–2875 (2015).

    Article  Google Scholar 

  25. Varela, L. & Horvath, T. L. Leptin and insulin pathways in POMC and AgRP neurons that modulate energy balance and glucose homeostasis. EMBO Rep. 13, 1079–1086 (2012).

    Article  CAS  Google Scholar 

  26. Perino, A. & Schoonjans, K. TGR5 and immunometabolism: insights from physiology and pharmacology. Trends Pharmacol. Sci. 36, 847–857 (2015).

    Article  CAS  Google Scholar 

  27. Orskov, C., Poulsen, S. S., Møller, M. & Holst, J. J. Glucagon-like peptide-1 receptors in the subfornical organ and the area postrema are accessible to circulating glucagon-like peptide-1. Diabetes 45, 832–835 (1996).

    Article  CAS  Google Scholar 

  28. Yamamoto, H. et al. Glucagon-like peptide-1-responsive catecholamine neurons in the area postrema link peripheral glucagon-like peptide-1 with central autonomic control sites. J. Neurosci. 23, 2939–2946 (2003).

    Article  CAS  Google Scholar 

  29. Rüttimann, E. B., Arnold, M., Hillebrand, J. J., Geary, N. & Langhans, W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology 150, 1174–1181 (2009).

    Article  Google Scholar 

  30. Wu, X. et al. Satiety induced by bile acids is mediated via vagal afferent pathways. JCI Insight 5, e132400 (2020).

    Article  Google Scholar 

  31. Pinto, S. et al. Rapid rewiring of arcuate nucleus feeding circuits by leptin. Science 304, 110–115 (2004).

    Article  CAS  Google Scholar 

  32. Zhu, Y. et al. Ablation of NF1 function in neurons induces abnormal development of cerebral cortex and reactive gliosis in the brain. Genes Dev. 15, 859–876 (2001).

    Article  CAS  Google Scholar 

  33. Tong, Q., Ye, C.-P., Jones, J. E., Elmquist, J. K. & Lowell, B. B. Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat. Neurosci. 11, 998–1000 (2008).

    Article  CAS  Google Scholar 

  34. Balthasar, N. et al. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 42, 983–991 (2004).

    Article  CAS  Google Scholar 

  35. Argmann, C. A., Champy, M.-F. & Auwerx, J. Evaluation of energy homeostasis. Curr. Protoc. Mol. Biol. 29, 29B.1 (2006).

    Google Scholar 

  36. Paxinos, G. & Franklin, K. B. J. The Mouse Brain in Stereotaxic Coordinates: Compact Second Edition (Academic Press, 2003).

  37. Bellocchio, L. et al. Activation of the sympathetic nervous system mediates hypophagic and anxiety-like effects of CB1 receptor blockade. Proc. Natl Acad. Sci. USA 110, 4786–4791 (2013).

    Article  CAS  Google Scholar 

  38. Saucisse, N. et al. POMC neurons functional heterogeneity relies on mTORC1 signaling. Preprint at bioRxiv http://biorxiv.org/lookup/doi/10.1101/2020.03.25.007765 (2020).

  39. Sorrentino, G. et al. Bile acids signal via TGR5 to activate intestinal stem cells and epithelial regeneration. Gastroenterology 159, 956–968.e8 (2020).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Fouassier, S. Bichet, T. Clerc, P. Colin, A. Aebi, T. Teav (Metabolomics Platform), the Histology facility and the Phenotyping Unit (UDP) for technical assistance, the Mouse Clinical Institute in Strasbourg for the generation of the TGR5:GFP mouse strain, and the Bordeaux Imaging Center (CNRS-INSERM and Bordeaux University; ANR-10-INBS-04) for confocal microscopy. This work was funded by the Swiss National Science Foundation (SNSF nos. 310030_189178 and CRSII5_180317/1, the Kristian Gerhard Jebsen Foundation and the Ecole Polytechnique Fédérale de Lausanne (EPFL; to K.S.), the National Institutes of Health (DK097566 to S.D.) and INSERM, ANR-17-CE14-0007, ANR-10-EQX-008-1 OPTOPATH (to D.C.). L.A.V.-V. and A.P. were supported by a postdoctoral fellowship from the AXA Research Fund.

Author information

Authors and Affiliations

Authors

Contributions

A.P., L.A.V.-V. and K.S. conceived and designed the project. B.S., S.D. and D.C. provided critical expertise. A.G. and R.P. provided chemical compounds. A.P., L.A.V.-V., N.B., Y.S., Q.H., V.F., A.C.-J., P.Z., G.B., S.J. and A.B. performed experiments. A.P., L.A.V.-V., N.B., V.F., A.C.-J., P.Z., G.B., S.J. and J.I. analysed the data. A.P., L.A.V.-V. and K.S. wrote the manuscript. All the authors read and approved the manuscript.

Corresponding author

Correspondence to Kristina Schoonjans.

Ethics declarations

Competing interests

A.G. and R.P. are cofounders of TES Pharma. R.P. is the president and CEO of TES Pharma. The other authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Richard Palmiter and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Christoph Schmitt.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 BA mix and INT-777 reach the hypothalamus after oral or icv administration, respectively, and reduce food intake by activating TGR5.

a, Conjugated and unconjugated BA species measured in the hypothalamus 30- and 60-min after oral administration of a BA mix at three different doses or vehicle in wild-type mice (n = 8 animals). Bars represent the mean from 8 replicates. b, One-hour cumulative food intake of wild-type mice after oral administration of a BA mix at three different doses or vehicle. n = 8 (Vehicle and 1mg/Kg) and n = 10 (10mg/Kg and 100mg/Kg) animals. c, Gpbar1 mRNA levels in arcuate nucleus (ARC)-enriched hypothalamic punches of TGR5 wild-type (Gpbar1+/+) and germline TGR5 knockout (Gpbar1-/-) mice. n = 8 animals. d, One-hour (1 h) food intake measured after the first i.c.v injection (d1 – 1 h) of vehicle (n = 8 animals) or INT-777 (n = 9 animals) in wild-type mice. e, Two-hour (2 h) food intake measured after the first i.c.v injection (d1 – 2 h) of vehicle (n = 8 animals) or INT-777 (n = 9 animals) in wild-type mice. f, Food intake measured after a daily (right before the dark phase) i.c.v injection of vehicle (n = 8 animals) or INT-777 (n = 9 animals) in wild-type mice at the indicated time points. g, Body weight change (expressed as a percentage of the initial body weight) of vehicle (n = 8 animals) or INT-777 (n = 9 animals) in wild-type mice at the indicated time points. Results represent mean (a) or mean ± SEM (b-g). n represents biologically independent replicates. Two-tailed Student t-test (c, d and e) or two-way ANOVA followed by Bonferroni post-hoc correction (b) vs Vehicle (b, d-e) or Gpbar1+/+ (c) were used for statistical analysis. P values (exact value, * P ≤ 0.05 or **** P ≤ 0.0001) are indicated in the figure.

Source data

Extended Data Fig. 2 TGR5 activation in neurons reduces food intake.

a, Quantification of AgRP neurons in the arcuate nucleus of over-night fasted TGR5 wild-type (Gpbar1+/+, 4 animals) and TGR5 knockout (Gpbar1-/-, 3 animals) mice. b, Body weight curve of 8-week-old Gpbar1+/+ and Gpbar1-/- mice over an 8-week period. n = 8 animals. c, Body composition (lean and fat mass expressed as a percentage over the body weight (BW)) of Gpbar1+/+ and Gpbar1-/- mice. n = 10 animals. d, Validation of the TGR5:GFP knock-in mouse model (n = 4 animals) in which the mouse Gpbar1 gene was substituted with a construct containing the human GPBAR1, the F2a sequence and the Gfp. Brains from wild-type mice (n = 3 animals) were used as controls. e, Gpbar1 mRNA levels in arcuate nucleus (ARC)-enriched hypothalamic punches of control mice (Gpbar1fl/fl) and neuron-specific TGR5 knockout (SynCre;Gpbar1fl/fl) mice. n = 8 animals. f, Body weight curve of 8-week-old of control mice (Gpbar1fl/fl, 10 animals) and neuron-specific TGR5 knockout (SynCre;Gpbar1fl/fl, 9 animals) mice over an 8-week period. g, Body composition (lean and fat mass expressed as percentage over body weight (BW)) of Gpbar1fl/fl (n = 10 animals) and SynCre;Gpbar1fl/fl (n = 6 animals) mice. h, 12-hour food intake of Gpbar1fl/fl (n = 7 animals) and SynCre;Gpbar1fl/fl (n = 8 animals) mice. i, Pomc mRNA levels in ARC-enriched hypothalamic punches of Gpbar1fl/fl and SynCre;Gpbar1fl/fl mice. n = 7 (SynCre;Gpbar1fl/fl fasting) and n = 8 (all other groups) animals. j, 12-hour cumulative food intake of control mice (Gpbar1fl/fl) and neuron-specific TGR5 knockout (SynCre;Gpbar1fl/fl) mice after oral administration of INT-777 (100 mg/Kg) or vehicle. n = 5 (Gpbar1fl/fl), n = 8 (SynCre;Gpbar1fl/fl INT-777) and n = 9 (SynCre;Gpbar1fl/fl vehicle) animals. k, Total food intake of Gpbar1fl/fl and SynCre;Gpbar1fl/fl during night and day. n = 6 (Gpbar1fl/fl INT-777 night), n = 7 (Gpbar1fl/fl other groups), n = 8 (SynCre;Gpbar1fl/fl other groups) and n = 9 (SynCre;Gpbar1fl/fl vehicle day) animals. l, 12-hour cumulative food intake of control mice (Gpbar1+/+) and germline TGR5 knock-out (Gpbar1-/-) mice after oral administration of the TGR5 specific BA agonist INT-777 (100 mg/Kg) or vehicle. n = 6 (Gpbar1+/+ vehicle), n = 7 (Gpbar1-/- vehicle) and n = 8 (all other groups) animals. m, Total food intake of Gpbar1+/+ and Gpbar1-/- mice during night and day. n = 7 (Gpbar1-/- vehicle night) and n = 8 (all other groups) animals. Results represent mean ± SEM. n represents biologically independent replicates. Two-tailed Student t-test (d, e, h and i), one-way ANOVA (k and m) or two-way ANOVA (j and l) followed by Bonferroni post-hoc corrections vs. Wild-type (d), Gpbar1fl/fl (e, h and i), Gpbar1fl/fl Vehicle (j and k) or Gpbar1+/+ Vehicle (l and m) were used for statistical analysis. P values (exact value, * P ≤ 0.05 or **** P ≤ 0.0001) are indicated in the figure.

Source data

Extended Data Fig. 3 Disruption of TGR5 in AgRP or POMC neurons in CD-fed mice does not change body weight and composition.

a, Gpbar1 mRNA levels in arcuate nucleus (ARC)-enriched hypothalamic punches of control mice (Gpbar1fl/fl) and POMC neuron-specific TGR5 knockout (PomcCre;Gpbar1fl/fl) mice. n = 6 animals. b, 12-hours food intake of Gpbar1fl/fl and PomcCre;Gpbar1fl/fl mice. n = 7 animals. c, Body weight curve of 8-week-old of control mice (Gpbar1fl/fl, n = 7 animals) and AgRP neuron-specific TGR5 knockout (AgRPCre;Gpbar1fl/fl, n = 9 animals) mice over an 8-week period. d, Body composition (lean and fat mass expressed as percentage over body weight (BW)) of Gpbar1fl/fl and AgRPCre;Gpbar1fl/fl mice. n = 8 animals. e, Body weight curve of 8-week-old of control mice (Gpbar1fl/fl) and POMC neuron-specific TGR5 knockout (PomcCre;Gpbar1fl/fl) mice over an 8-week period. n = 10 animals. f, Body composition (lean and fat mass expressed as percentage over body weight (BW)) of Gpbar1fl/fl (n = 7 animals) and PomcCre;Gpbar1fl/fl (n = 8 animals) mice. g-h, Representative images (g) and quantification (h) of Fos immunoreactivity (in red) in AgRP/NPY neurons (in green) and merge (in orange) in the hypothalamus of fed or fasted AgRP/NPY:GFP reporter mice. Arrows in images indicate colocalization of Fos and GFP. Scale bar = 100 μm. n = 4 animals. Results represent mean ± SEM. n represents biologically independent replicates. Two-tailed Student t-test vs. Gpbar1fl/fl (a) or fed AgRP/NPY-GFP (h) groups was used for statistical analysis. P values are indicated in the figure.

Source data

Extended Data Fig. 4 TGR5 inhibits AgRP secretion through activation of the Rho/ROCK/actin signalling axis.

a, Gamma-aminobutyric acid (GABA) release in ex vivo hypothalamic explants from TGR5 wild-type (Gpbar1+/+) and germline TGR5 knockout (Gpbar1-/-) mice after starvation or starvation followed by 10- and 30-minutes stimulation with TGR5 agonist INT-777 or vehicle. n = 4 (all other groups) and n = 8 (Gpbar1+/+ vehicle 10min) animals. b-c, GABA (b) or alpha-melanocyte-stimulating hormone (αMSH) (c) release in ex vivo hypothalamic explants from Gpbar1+/+ and Gpbar1-/- mice after high-glucose (15 mM) solution to mimic fed conditions, followed by 10- and 30-minutes stimulation with TGR5 agonist INT-777 or vehicle. n = 4 (b), n = 4 (all other groups in c) and n = 8 (Gpbar1+/+ and Gpbar1-/- fed, c) animals. d, Gpbar1 mRNA levels in mouse embryonic hypothalamic N41 cell line (mHypoE-N41) after Gpbar1 silencing using transient transfection of sh-Gpbar1 or control vector (sh-Ctrl). n = 12 samples. e, Agrp and Npy mRNA levels in mHypoE-N41 cells after 60 min starvation conditions followed by 60 min stimulation with INT-777 or vehicle. n = 3 samples. f, AgRP release after starvation followed by short-term (5, 10 and 15 min) stimulation with TGR5 agonist INT-777 or vehicle in mHypoE-N41 cells. n = 3 samples. g, AgRP release after starvation followed by 10 min stimulation with INT-777 or vehicle in mHypoE-N41 Gpbar1-silenced cells using transient transfection of sh-Gpbar1 or control vector (sh-Ctrl). n = 3 samples. (h and i) Quantitative densitometry of phosphorylated vs total ROCK signalling targets (MLC and COFILIN) from the cells described in Fig. 4f. n = 4 samples. j, Representative images of phalloidin staining to detect actin fibres (in green) and DAPI staining to detect nuclei (in blue), phosphorylated COFILIN (p-COFILIN) immunodetection (in red) and merge, after 10 min stimulation with INT-777, BA mix or vehicle in mHypoE-N41 cells. Scale bar = 10μm and digital zoom. n = 4 samples. Results represent mean ± SEM. n represents biologically independent replicates. Two-tailed Student t-test (d-f) or one-way ANOVA followed by Bonferroni post-hoc correction (g, h and i) vs. sh-Ctrl (d) or vehicle (e-i) groups were used for statistical analysis. P values (exact value or **** P ≤ 0.0001) are indicated in the figure.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Elution gradient for chromatographic separation of BAs.

Supplementary Table 2

Oligonucleotide sequences.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed and uncropped western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Perino, A., Velázquez-Villegas, L.A., Bresciani, N. et al. Central anorexigenic actions of bile acids are mediated by TGR5. Nat Metab 3, 595–603 (2021). https://doi.org/10.1038/s42255-021-00398-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00398-4

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