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.

  • Article
  • Published:

Secretin activates brown fat and induces satiation

Abstract

Brown adipose tissue (BAT) thermogenesis is activated by feeding. Recently, we revealed a secretin-mediated gut–BAT–brain axis, which stimulates satiation in mice, but the purpose of meal-induced BAT activation in humans has been unclear. In this placebo-controlled, randomized crossover study, we investigated the effects of intravenous secretin on BAT metabolism (measured with [18F]FDG and [15O]H2O positron emission tomography) and appetite (measured with functional magnetic resonance imaging) in healthy, normal weight men (GUTBAT trial no. NCT03290846). Participants were blinded to the intervention. Secretin increased BAT glucose uptake (primary endpoint) compared to placebo by 57% (median (interquartile range, IQR), 0.82 (0.77) versus 0.59 (0.53) μmol per 100 g per min, 95% confidence interval (CI) (0.09, 0.89), P = 0.002, effect size r = 0.570), while BAT perfusion remained unchanged (mean (s.d.) 4.73 (1.82) versus 6.14 (3.05) ml per 100 g per min, 95%CI (−2.91, 0.07), P = 0.063, effect size d = −0.549) (n = 15). Whole body energy expenditure increased by 2% (P = 0.011) (n = 15). Secretin attenuated blood-oxygen level-dependent activity (primary endpoint) in brain reward circuits during food cue tasks (significance level false discovery rate corrected at P = 0.05) (n = 14). Caloric intake did not significantly change, but motivation to refeed after a meal was delayed by 39 min (P = 0.039) (n = 14). No adverse effects were detected. Here we show in humans that secretin activates BAT, reduces central responses to appetizing food and delays the motivation to refeed after a meal. This suggests that meal-induced, secretin-mediated BAT activation is relevant in the control of food intake in humans. As obesity is increasing worldwide, this appetite regulating axis offers new possibilities for clinical research in treating obesity.

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: Study protocol.
Fig. 2: Secretin increases GU in human BAT.
Fig. 3: Secretin induces satiation when measured by fMRI.
Fig. 4: Secretin induces satiation when measured by CSS.
Fig. 5: SCTR expression in BAT negatively correlates with body weight in fasted participants.
Fig. 6: Secretin changes circulating metabolite and hormone levels.

Similar content being viewed by others

Data availability

All statistical parametric images of the fMRI study (including the effect size maps) can be found from NeuroVault at https://neurovault.org/collections/ECURNRON/. The accession number for the RNA-Seq data presented in this article is GEO GSE113764. SCTR expression in human tissues is available in the Online Biology Gene Portal System (BioGPS) at http://ds.biogps.org/?dataset=GSE1133&gene=6344. Datasets that support the findings of this study are available in the Supplementary Information. Source data are provided with this paper. The data that support the plots within this paper, as well as other findings of this study, are available from the corresponding author (P.N.) upon reasonable request.

References

  1. Virtanen, K. A. et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 360, 1518–1525 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Cypess, A. M. et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 360, 1509–1517 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. van Marken Lichtenbelt, W. D. et al. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 360, 1500–1508 (2009).

    Article  PubMed  Google Scholar 

  4. Blondin, D. P. et al. Contributions of white and brown adipose tissues and skeletal muscles to acute cold-induced metabolic responses in healthy men. J. Physiol. 593, 701–714 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Villarroya, F., Cereijo, R., Villarroya, J. & Giralt, M. Brown adipose tissue as a secretory organ. Nat. Rev. Endocrinol. 13, 26–35 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Kajimura, S., Spiegelman, B. M. & Seale, P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab. 22, 546–559 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rothwell, N. J. & Stock, M. J. A role for brown adipose tissue in diet-induced thermogenesis. Nature 281, 31–35 (1979).

    Article  CAS  PubMed  Google Scholar 

  8. Glick, Z., Teague, R. J. & Bray, G. A. Brown adipose tissue: thermic response increased by a single low protein, high carbohydrate meal. Science 213, 1125–1127 (1981).

    Article  CAS  PubMed  Google Scholar 

  9. U Din, M. et al. Postprandial oxidative metabolism of human brown fat indicates thermogenesis. Cell Metab. 28, 207–216.e3 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Chondronikola, M., Porter, C., Malagaris, I., Nella, A. A. & Sidossis, L. S. Brown adipose tissue is associated with systemic concentrations of peptides secreted from the gastrointestinal system and involved in appetite regulation. Eur. J. Endocrinol. 177, 33–40 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Crovetti, R., Porrini, M., Santangelo, A. & Testolin, G. The influence of thermic effect of food on satiety. Eur. J. Clin. Nutr. 52, 482–488 (1998).

    Article  CAS  PubMed  Google Scholar 

  12. Li, Y. et al. Secretin-activated brown fat mediates prandial thermogenesis to induce satiation. Cell 175, 1561–1574.e12 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Blondin, D. et al. Inhibition of intracellular triglyceride lipolysis suppresses cold-induced brown adipose tissue metabolism and increases shivering in humans. Cell Metab. 25, 438–447 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Wu, C., Jin, X., Tsueng, G., Afrasiabi, C. & Su, A. I. BioGPS: building your own mash-up of gene annotations and expression profiles. Nucleic Acids Res. 44, D313–D316 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Su, A. I. et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proc. Natl Acad. Sci. USA. 101, 6062–6067 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nummenmaa, L. et al. μ-opioid receptor system mediates reward processing in humans. Nat. Commun. 9, 1500 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Gilbert, J.-A. et al. The effect of tesofensine on appetite sensations. Obesity 20, 553–561 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Balaz, M. et al. Inhibition of mevalonate pathway prevents adipocyte browning in mice and men by affecting protein prenylation. Cell Metab. 29, 901–916.e8 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Sekar, R. & Chow, B. K. C. Lipolytic actions of secretin in mouse adipocytes. J. Lipid Res. 55, 190–200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Butcher, R. W. & Carlson, L. A. Effects of secretin on fat mobilizing lipolysis and cyclic AMP levels in rat adipose tissue. Acta Physiol. Scand. 79, 559–563 (1970).

    Article  CAS  PubMed  Google Scholar 

  21. Raiko, J. et al. Brown adipose tissue triglyceride content is associated with decreased insulin sensitivity, independently of age and obesity. Diabetes, Obes. Metab. 17, 516–519 (2015).

    Article  CAS  Google Scholar 

  22. U Din, M. et al. Human brown fat radiodensity indicates underlying tissue composition and systemic metabolic health. J. Clin. Endocrinol. Metab. 102, 2258–2267 (2017).

    Article  PubMed  Google Scholar 

  23. Yaney, G. C. & Corkey, B. E. Fatty acid metabolism and insulin secretion in pancreatic beta cells. Diabetologia 46, 1297–1312 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Stein, D. T. et al. The insulinotropic potency of fatty acids is influenced profoundly by their chain length and degree of saturation. J. Clin. Invest. 100, 398–403 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Afroze, S. et al. The physiological roles of secretin and its receptor. Ann. Transl. Med. 1, 29 (2013).

    PubMed  PubMed Central  Google Scholar 

  26. Broeders, E. P. M. et al. The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab. 22, 418–426 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Amin, A., Dhillo, W. S. & Murphy, K. G. The central effects of thyroid hormones on appetite. J. Thyroid Res. 2011, 306510 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Bianco, A. C. & Silva, J. E. Intracellular conversion of thyroxine to triiodothyronine is required for the optimal thermogenic function of brown adipose tissue. J. Clin. Invest. 79, 295–300 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lahesmaa, M. et al. Hyperthyroidism increases brown fat metabolism in humans. J. Clin. Endocrinol. Metab. 99, E28–E35 (2014).

    Article  PubMed  Google Scholar 

  30. Bessesen, D. H. & Van Gaal, L. F. Progress and challenges in anti-obesity pharmacotherapy. Lancet Diabetes Endocrinol. 6, 237–248 (2018).

    Article  PubMed  Google Scholar 

  31. van Ooijen, A. M. J., van Marken Lichtenbelt, W. D., van Steenhoven, A. A. & Westerterp, K. R. Seasonal changes in metabolic and temperature responses to cold air in humans. Physiol. Behav. 82, 545–553 (2004).

    Article  PubMed  Google Scholar 

  32. Yoneshiro, T. et al. Recruited brown adipose tissue as an antiobesity agent in humans. J. Clin. Invest. 123, 3404–3408 (2013).

  33. U.-Din, M. et al. Human brown adipose tissue [15O]O2 PET imaging in the presence and absence of cold stimulus. Eur. J. Nucl. Med. Mol. Imag. https://doi.org/10.1007/s00259-016-3364-y (2016).

  34. Orava, J. et al. Different metabolic responses of human brown adipose tissue to activation by cold and insulin. Cell Metab. 14, 272–279 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Patlak, C. S. & Blasberg, R. G. Graphical evaluation of blood-to-brain transfer constants from multiple-time uptake data. Generalizations. J. Cereb. Blood Flow. Metab. 5, 584–590 (1985).

    Article  CAS  PubMed  Google Scholar 

  36. Virtanen, K. A. et al. Human adipose tissue glucose uptake determined using [18 F]-fluoro-deoxy-glucose ([18 F]FDG) and PET in combination with microdialysis. Diabetologia 44, 2171–2179 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Peltoniemi, P. et al. Lumped constant for [18 F]fluorodeoxyglucose in skeletal muscles of obese and nonobese humans. Am. J. Physiol. Metab. 279, E1122–E1130 (2000).

    CAS  Google Scholar 

  38. Weir, J. B. & de, V. New methods for calculating metabolic rate with special reference to protein metabolism. J. Physiol. 109, 1–9 (1949).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Meriläinen, P. T. Metabolic monitor. Int. J. Clin. Monit. Comput. 4, 167–177 (1987).

    Article  PubMed  Google Scholar 

  40. Nummenmaa, L. et al. Dorsal striatum and its limbic connectivity mediate abnormal anticipatory reward processing in obesity. PLoS ONE 7, e31089 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Esteban, O. et al. fMRIPrep: a robust preprocessing pipeline for functional MRI. Nat. Methods https://doi.org/10.1038/s41592-018-0235-4 (2019).

  42. Gorgolewski, K. et al. Nipype: A flexible, lightweight and extensible neuroimaging data processing framework in Python. Front. Neuroinform. https://doi.org/10.3389/fninf.2011.00013 (2011).

  43. Tustison, N. J. et al. N4ITK: Improved N3 bias correction. IEEE Trans. Med. Imag. https://doi.org/10.1109/TMI.2010.2046908 (2010).

  44. Dale, A. M., Fischl, B. & Sereno, M. I. Cortical surface-based analysis: I. Segmentation and surface reconstruction. Neuroimage https://doi.org/10.1006/nimg.1998.0395 (1999).

  45. Klein, A. et al. Mindboggling morphometry of human brains. PLoS Comput. Biol. https://doi.org/10.1371/journal.pcbi.1005350 (2017).

  46. Fonov, V., Evans, A., McKinstry, R., Almli, C. & Collins, D. Unbiased nonlinear average age-appropriate brain templates from birth to adulthood. Neuroimage https://doi.org/10.1016/s1053-8119(09)70884-5 (2009).

  47. Avants, B. B., Epstein, C. L., Grossman, M. & Gee, J. C. Symmetric diffeomorphic image registration with cross-correlation: evaluating automated labeling of elderly and neurodegenerative brain. Med. Image Anal. https://doi.org/10.1016/j.media.2007.06.004 (2008).

  48. Zhang, Y., Brady, M. & Smith, S. Segmentation of brain MR images through a hidden Markov random field model and the expectation-maximization algorithm. IEEE Trans. Med. Imag. https://doi.org/10.1109/42.906424 (2001).

  49. Cox, R. W. AFNI: Software for analysis and visualization of functional magnetic resonance neuroimages. Comput. Biomed. Res. https://doi.org/10.1006/cbmr.1996.0014 (1996).

  50. Jenkinson, M., Bannister, P., Brady, M. & Smith, S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage https://doi.org/10.1016/S1053-8119(02)91132-8 (2002).

  51. Greve, D. N. & Fischl, B. Accurate and robust brain image alignment using boundary-based registration. Neuroimage https://doi.org/10.1016/j.neuroimage.2009.06.060 (2009).

  52. Behzadi, Y., Restom, K., Liau, J. & Liu, T. T. A component based noise correction method (CompCor) for BOLD and perfusion based fMRI. Neuroimage https://doi.org/10.1016/j.neuroimage.2007.04.042 (2007).

  53. Power, J. D. et al. Methods to detect, characterize, and remove motion artifact in resting state fMRI. Neuroimage https://doi.org/10.1016/j.neuroimage.2013.08.048 (2014).

  54. Pruim, R. H. R. et al. ICA-AROMA: a robust ICA-based strategy for removing motion artifacts from fMRI data. Neuroimage https://doi.org/10.1016/j.neuroimage.2015.02.064 (2015).

  55. Flint, A., Raben, A., Blundell, J. E. & Astrup, A. Reproducibility, power and validity of visual analogue scales in assessment of appetite sensations in single test meal studies. Int. J. Obes. Relat. Metab. Disord. 24, 38–48 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Soininen, P. et al. High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst 134, 1781 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Li, Y., Fromme, T., Schweizer, S., Schöttl, T. & Klingenspor, M. Taking control over intracellular fatty acid levels is essential for the analysis of thermogenic function in cultured primary brown and brite/beige adipocytes. EMBO Rep. 15, 1069–1076 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Rosenthal, R. in The Handbook of Research Synthesis (eds Cooper, H. et al.) 231–244 (Russell Sage Foundation, 1994).

  59. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B-Methodol. 57, 289–300 (1995).

    Google Scholar 

Download references

Acknowledgements

The study was conducted within the Centre of Excellence into Cardiovascular and Metabolic Diseases supported by the Academy of Finland (grant no. 307402), University of Turku, Åbo Akademi University; and funded by the Instrumentarium Science Foundation (grant no. 190014) (S.L.), The Paulo Foundation (S.L.), Turku University Hospital Foundation (S.L.) and The Finnish Medical Foundation (grant no. 2985) (S.L.). TUM researchers were supported by the Else Kröner-Fresenius Stiftung (M.K.) and the German Research Foundation (grant no. DFG-CRC 1371: P13 (M.K.), Z02 (K. Steiger)).

Author information

Authors and Affiliations

Authors

Contributions

M.K., P.N., K.A.V. and S.L. conceived and designed the study. S.L., L.S. and M.L. performed the human experiments. K. Schnabl conceived the study design for the SCTR expression and regulation study and carried out animal and cell culture experiments. S.L., L.S., K. Schnabl, K.L., M.U.-D. and R.K analysed data. L.N., L.L.E. and T.V. helped with statistical analysis. O.E. and A.K.K. contributed to PET/CT data collection. K. Steiger conducted the immunohistochemical studies. L.N. conceived the fMRI study design. T.N. and M.T. provided BAT biopsies from study participants. M.B. and C.W. provided the analysis of fasted human BAT biopsies. K. Schnabl, Y.L and M.K edited the paper and contributed physiological aspects of secretin-activated brown fat in mouse studies complementing the human study. S.L. wrote the paper with input from authors. All authors read and approved the paper.

Corresponding author

Correspondence to Pirjo Nuutila.

Ethics declarations

Competing interests

M.K. and Y.L. are inventors on a patent application from the Technical University of Munich (publication no. WO/2017/20285; international application no. PCT/EP2017/062420) addressing the role of SCTR agonists and modulators in the regulation of energy homeostasis. This patent is based on the initial discovery that meal-induced secretin inhibits food intake, and this anorexigenic action of secretin depends on the activation of brown fat12. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Andrew Gray, Marc Tittgemeyer 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 Enrolment and analysis.

CONSORT flowchart on the enrolment and analysis of study participants.

Extended Data Fig. 2 Visual analogue scale –questions.

Composite satiety score calculated with scale 1–100. Composite satiety score = satiety + fullness + (100 − prospective food consumption) + (100 − hunger)).

Extended Data Fig. 3 PET results and secretin receptor expression.

a. Muscle glucose uptake (GU) is increased by secretin compared to placebo. Data were analyzed with two-sided paired samples t-test and included n = 15 subjects. b. Secretin receptor expression in 79 human tissues, retrieved from http://ds.biogps.org/?dataset=GSE1133&gene=6344. Data are expressed as fold over the median (M), error bars show standard error. c. There is no significant change in perfusion after secretin infusion as compared to placebo. Data were analyzed with two-sided paired samples t-test and included n = 15 subjects.

Source data

Extended Data Fig. 4 Additional functional magnetic resonance and composite satiety score results.

a, Down-regulated brain reward anticipation response due to injection of secretin. This separate analysis (n = 14) including subjects with high movement artifacts was originally done. In the placebo condition, palatable food led to increased BOLD activity in brain reward circuits. This effect was diminished in the secretin condition. Interaction effect between food categories and conditions were found in the reward circuits. Significance level was FDR-corrected at p value 0.05. MFC = medial frontal cortex, CC = cingulate cortex, OCC = occipital cortex, Cau = caudate, MTG = middle temporal gyrus, Ins = insula, PCC = posterior cingulate cortex. b, composite satiety score area under the curve (AUC) was not increased postprandially, c nor for the entire study day (pre-prandial, prandial and postprandial phases together). Values are normalized, dividing by the value of the first time point. Data were analyzed by two-sided paired samples t-test, n = 14.

Source data

Extended Data Fig. 5 Additional positron emission tomography and secretin receptor gene expression results.

a, Brown adipose tissue (BAT) glucose uptake (GU) in cold and after secretin. There is no correlation between BAT GU in cold exposure and after secretin infusion. Data were analyzed by Spearman’s rank correlation in order to avoid assumptions around linearity of associations (n = 15). b, human secretin receptor (SCTR) expression on mRNA level, assessed by qPCR, in supraclavicular BAT is weakly associated with body-mass-index (BMI) in n = 14 fasted participants. Data were analyzed by Pearson’s correlation. c, SCTR expression analyzed by RNASeq does not correlate with BMI in n = 14 non-fasted participants. Data were analyzed by Pearson’s correlation.

Source data

Extended Data Fig. 6 Changes in circulating bile acids, hormones and carbohydrates.

a. Secretin did not increase serum bile acid levels compared to placebo. Values are normalized, dividing by the value of the first time point. Each timepoint was analyzed by two-sided, paired Wilcoxon signed-rank test. Median values and standard errors are shown on graph, n = 14. b. Serum hormone and c. carbohydrate heatmap secretin vs. placebo. Values are normalized, dividing by the value of the first time point. Each timepoint was analyzed by two-sided paired Wilcoxon signed-rank test, n = 12. * = p < 0.05, ** = p < 0.01, *** p < 0.001.

Source data

Supplementary information

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Laurila, S., Sun, L., Lahesmaa, M. et al. Secretin activates brown fat and induces satiation. Nat Metab 3, 798–809 (2021). https://doi.org/10.1038/s42255-021-00409-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00409-4

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