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:

Central and peripheral GLP-1 systems independently suppress eating

Subjects

Abstract

The anorexigenic peptide glucagon-like peptide-1 (GLP-1) is secreted from gut enteroendocrine cells and brain preproglucagon (PPG) neurons, which, respectively, define the peripheral and central GLP-1 systems. PPG neurons in the nucleus tractus solitarii (NTS) are widely assumed to link the peripheral and central GLP-1 systems in a unified gut–brain satiation circuit. However, direct evidence for this hypothesis is lacking, and the necessary circuitry remains to be demonstrated. Here we show that PPGNTS neurons encode satiation in mice, consistent with vagal signalling of gastrointestinal distension. However, PPGNTS neurons predominantly receive vagal input from oxytocin-receptor-expressing vagal neurons, rather than those expressing GLP-1 receptors. PPGNTS neurons are not necessary for eating suppression by GLP-1 receptor agonists, and concurrent PPGNTS neuron activation suppresses eating more potently than semaglutide alone. We conclude that central and peripheral GLP-1 systems suppress eating via independent gut–brain circuits, providing a rationale for pharmacological activation of PPGNTS neurons in combination with GLP-1 receptor agonists as an obesity treatment strategy.

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

Fig. 1: PPGNTS neurons selectively encode large meal satiation.
Fig. 2: PPGNTS neurons suppress eating without behavioural disruption.
Fig. 3: Glp1r-expressing VANs suppress eating and condition flavour avoidance.
Fig. 4: Oxtr rather than Glp1r VANs are the major vagal input to PPGNTS neurons.
Fig. 5: PPGNTS neurons are necessary for oxytocin-induced eating suppression.
Fig. 6: PPGNTS neurons are not a major target of AP Glp1r neurons.
Fig. 7: Liraglutide and semaglutide suppress eating independently of PPGNTS neurons.
Fig. 8: PPGNTS neuron activation augments semaglutide-induced eating suppression.

Similar content being viewed by others

Data availability

There are no publicly available datasets for this manuscript. All data necessary to interpret, replicate and build on the methods or findings reported here are contained within the manuscript and extended data figures. Primary data from ex vivo calcium recordings, food intake, metabolic and behavioural analyses, videos for BSS coding and photomicrographs for in situ hybridization and immunohistochemistry analyses are available in native format upon request from the corresponding authors. Access to stored tissue samples used for in situ hybridization and immunohistochemistry is available upon request from the corresponding authors. All mouse lines, plasmids and reagents used in this study have been previously published and/or are commercially available, and are detailed in the Reporting Summary. Further information and requests for resources and reagents should be directed to and will be fulfilled by S. Trapp (s.trapp@ucl.ac.uk).

References

  1. Knudsen, L. B. & Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. 10, 155 (2019).

    Article  Google Scholar 

  2. Turton, M. D. et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 69–72 (1996).

    Article  CAS  PubMed  Google Scholar 

  3. Trapp, S. & Richards, J. E. The gut hormone glucagon-like peptide-1 produced in brain: is this physiologically relevant? Curr. Opin. Pharmacol. 13, 964–969 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Müller, T. D. et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 30, 72–130 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Gaykema, R. P. et al. Activation of murine pre-proglucagon–producing neurons reduces food intake and body weight. J. Clin. Invest. 127, 1031–1045 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Liu, J. et al. Enhanced AMPA Receptor trafficking mediates the anorexigenic effect of endogenous glucagon-like peptide-1 in the paraventricular hypothalamus. Neuron 96, 897–909.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Holt, M. K. et al. Preproglucagon neurons in the nucleus of the solitary tract are the main source of brain GLP-1, mediate stress-induced hypophagia, and limit unusually large intakes of food. Diabetes 68, 21–33 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Zheng, H., Stornetta, R. L., Agassandian, K. & Rinaman, L. Glutamatergic phenotype of glucagon-like peptide 1 neurons in the caudal nucleus of the solitary tract in rats. Brain Struct. Funct. 220, 3011–3022 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Trapp, S. et al. PPG neurons of the lower brain stem and their role in brain GLP-1 receptor activation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309, R795–R804 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cheng, W. et al. Leptin receptor–expressing nucleus tractus solitarius neurons suppress food intake independently of GLP1 in mice. JCI Insight 5, e134359 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Merchenthaler, I., Lane, M. & Shughrue, P. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J. Comp. Neurol. 403, 261–280 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Cork, S. C. et al. Distribution and characterisation of glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol. Metab. 4, 718–731 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Card, J. P. et al. GLP-1 neurons form a local synaptic circuit within the rodent nucleus of the solitary tract. J. Comp. Neurol. 526, 2149–2164 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hisadome, K., Reimann, F., Gribble, F. M. & Trapp, S. Leptin directly depolarizes preproglucagon neurons in the nucleus tractus solitarius: electrical properties of glucagon-like peptide 1 neurons. Diabetes 59, 1890–1898 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Drucker, D. J. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 27, 740–756 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Krieger, J. P. Intestinal glucagon-like peptide-1 effects on food intake: physiological relevance and emerging mechanisms. Peptides 131, 170342 (2020).

    Article  CAS  PubMed  Google Scholar 

  17. Grill, H. J. A role for GLP-1 in treating hyperphagia and obesity. Endocrinology 161, bqaa093 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Holt, M. K. et al. PPG neurons in the nucleus of the solitary tract modulate heart rate but do not mediate GLP-1 receptor agonist-induced tachycardia in mice. Mol. Metab. 39, 101024 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Secher, A. et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Invest. 124, 4473–4488 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gabery, S. et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight 5, e133429 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Varin, E. M. et al. Distinct neural sites of GLP-1R expression mediate physiological versus pharmacological control of incretin action. Cell Rep. 27, 3371–3384.e3 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Kanoski, S. E., Fortin, S. M., Arnold, M., Grill, H. J. & Hayes, M. R. Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology 152, 3103–3112 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Punjabi, M. et al. Circulating glucagon-like peptide-1 (GLP-1) inhibits eating in male rats by acting in the hindbrain and without inducing avoidance. Endocrinology 155, 1690–1699 (2014).

    Article  PubMed  Google Scholar 

  24. Ast, J. et al. Super-resolution microscopy compatible fluorescent probes reveal endogenous glucagon-like peptide-1 receptor distribution and dynamics. Nat. Commun. 11, 1–18 (2020).

    Google Scholar 

  25. Krieger, J. P. et al. Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes 65, 34–43 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Kim, K. S., Seeley, R. J. & Sandoval, D. A. Signalling from the periphery to the brain that regulates energy homeostasis. Nat. Rev. Neurosci. 19, 185–196 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Williams, E. K. K. et al. Sensory neurons that detect stretch and nutrients in the digestive system. Cell 166, 209–221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bai, L. et al. Genetic identification of vagal sensory neurons that control feeding. Cell 179, 1129–1143.e23 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Holt, M. K. et al. Synaptic inputs to the mouse dorsal vagal complex and its resident preproglucagon neurons. J. Neurosci. 39, 9767–9781 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kreisler, A. D., Davis, E. A. & Rinaman, L. Differential activation of chemically identified neurons in the caudal nucleus of the solitary tract in non-entrained rats after intake of satiating vs. non-satiating meals. Physiol. Behav. 136, 47–54 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Nguyen, K. P., O’Neal, T. J., Bolonduro, O. A., White, E. & Kravitz, A. V. Feeding experimentation device (FED): a flexible open-source device for measuring feeding behavior. J. Neurosci. Methods 267, 108–114 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kreisler, A. D. & Rinaman, L. Hindbrain glucagon-like peptide-1 neurons track intake volume and contribute to injection stress-induced hypophagia in meal-entrained rats. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 310, R906–R916 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Terrill, S. J. et al. Endogenous GLP-1 in lateral septum promotes satiety and suppresses motivation for food in mice. Physiol. Behav. 206, 191–199 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ishii, Y. et al. Differential effects of the selective orexin-1 receptor antagonist SB-334867 and lithium chloride on the behavioural satiety sequence in rats. Physiol. Behav. 81, 129–140 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Wright, F. L. & Rodgers, R. J. Behavioural profile of exendin-4/naltrexone dose combinations in male rats during tests of palatable food consumption. Psychopharmacology 231, 3729–3744 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Vrang, N., Phifer, C. B., Corkern, M. M. & Berthoud, H.-R. Gastric distension induces c-Fos in medullary GLP-1/2-containing neurons. Am. J. Physiol. Integr. Comp. Physiol. 285, R470–R478 (2003).

    Article  CAS  Google Scholar 

  37. Schwartz, G. J. & Moran, T. H. Sub-diaphragmatic vagal afferent integration of meal-related gastrointestinal signals. Neurosci. Biobehav. Rev. 20, 47–56 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Al Helaili, A., Park, S. J. & Beyak, M. J. Chronic high fat diet impairs glucagon like peptide-1 sensitivity in vagal afferents. Biochem. Biophys. Res. Commun. 533, 110–117 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Kanoski, S. E., Hayes, M. R. & Skibicka, K. P. GLP-1 and weight loss: unraveling the diverse neural circuitry. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 310, R885–R895 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Yamaguchi, E., Yasoshima, Y. & Shimura, T. Systemic administration of anorexic gut peptide hormones impairs hedonic-driven sucrose consumption in mice. Physiol. Behav. 171, 158–164 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Hayes, M. R. & Schmidt, H. D. GLP-influences food and drug reward. Curr. Opin. Behav. Sci. 9, 66–70 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Anesten, F. et al. Preproglucagon neurons in the hindbrain have IL-6 receptor-α and show Ca2+ influx in response to IL-6. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 311, R115–R123 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Holt, M. K., Llewellyn-Smith, I. J., Reimann, F., Gribble, F. M. & Trapp, S. Serotonergic modulation of the activity of GLP-1 producing neurons in the nucleus of the solitary tract in mouse. Mol. Metab. 6, 909–921 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Iwasaki, Y. et al. Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: a route for ameliorating hyperphagia and obesity. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 308, R360–R369 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Iwasaki, Y. et al. Relay of peripheral oxytocin to central oxytocin neurons via vagal afferents for regulating feeding. Biochem. Biophys. Res. Commun. 519, 553–558 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Kawatani, M., Yamada, Y. & Kawatani, M. Glucagon-like peptide-1 (GLP-1) action in the mouse area postrema neurons. Peptides 107, 68–74 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. 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  PubMed  PubMed Central  Google Scholar 

  48. Hisadome, K., Reimann, F., Gribble, F. M. & Trapp, S. CCK stimulation of GLP-1 neurons involves α 1-adrenoceptor- mediated increase in glutamatergic synaptic inputs. Diabetes 60, 2701–2709 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Adams, J. M. et al. Liraglutide modulates appetite and body weight through glucagon-like peptide 1 receptor-expressing glutamatergic neurons. Diabetes 67, 1538–1548 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Fortin, S. M. et al. GABA neurons in the nucleus tractus solitarius express GLP-1 receptors and mediate anorectic effects of liraglutide in rats. Sci. Transl. Med. 12, eaay8071 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Llewellyn-Smith, I. J., Reimann, F., Gribble, F. M. & Trapp, S. Preproglucagon neurons project widely to autonomic control areas in the mouse brain. Neuroscience 180, 111–121 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Reimann, F. et al. Glucose sensing in L cells: a primary cell study. Cell Metab. 8, 532–539 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Parker, H. E. et al. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia 55, 2445–2455 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Richards, P. et al. Identification and characterization of GLP-1 receptor-expressing cells using a new transgenic mouse model. Diabetes 63, 1224–1233 (2014).

    Article  CAS  PubMed  Google Scholar 

  55. Han, W. et al. A neural circuit for gut-induced reward. Cell 175, 665–678 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Rakipovski, G. et al. The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE−/− and LDLr−/− mice by a mechanism that includes inflammatory pathways. JACC Basic Transl. Sci. 3, 844–857 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  57. de Lartigue, G., Ronveaux, C. C. & Raybould, H. E. Deletion of leptin signaling in vagal afferent neurons results in hyperphagia and obesity. Mol. Metab. 3, 595–607 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Friard, O. & Gamba, M. BORIS: a free, versatile open-source event-logging software for video/audio coding and live observations. Methods Ecol. Evol. 7, 1325–1330 (2016).

    Article  Google Scholar 

  59. Halford, J. C., Wanninayake, S. C. & Blundell, J. E. Behavioral satiety sequence (BSS) for the diagnosis of drug action on food intake. Pharmacol. Biochem. Behav. 61, 159–168 (1998).

    Article  CAS  PubMed  Google Scholar 

  60. Rodgers, R. J., Holch, P. & Tallett, A. J. Behavioural satiety sequence (BSS): separating wheat from chaff in the behavioural pharmacology of appetite. Pharmacol. Biochem. Behav. 97, 3–14 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Mina, A. I. et al. CalR: a web-based analysis tool for indirect calorimetry experiments. Cell Metab. 28, 656–666.e1 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. de Kloet, A. D. et al. Reporter mouse strain provides a novel look at angiotensin type-2 receptor distribution in the central nervous system. Brain Struct. Funct. 221, 891–912 (2016).

    Article  PubMed  Google Scholar 

  63. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Wu, Z., Autry, A. E., Bergan, J. F., Watabe-Uchida, M. & Dulac, C. G. Galanin neurons in the medial preoptic area govern parental behaviour. Nature 509, 325–330 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Krashes, M. J. et al. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J. Clin. Invest. 121, 1424–1428 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Watabe-Uchida, M., Zhu, L., Ogawa, S. K., Vamanrao, A. & Uchida, N. Whole-brain mapping of direct inputs to midbrain dopamine neurons. Neuron 74, 858–873 (2012).

    Article  CAS  PubMed  Google Scholar 

  67. Wickersham, I. R. et al. Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron 53, 639–647 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L.B. Knudsen at Novo Nordisk for valuable discussions and provision of liraglutide and semaglutide. We also thank M. Arnold at ETH Zurich and Y. Tan at the University of Florida for their expert technical assistance, and K. Beier at UC Irvine for providing RABV for retrograde tracing. This study was supported by Medical Research Council (MRC) project grant no. MR/N02589X/1 to S.T. The initial collaboration between the Trapp, de Lartigue and Langhans laboratories, which generated this work was made possible thanks to a UCL Global Engagement Fund award to D.I.B., and a UCL Neuroscience ZNZ Collaboration award to S.T. and W.L. Research in the de Lartigue laboratory was funded by the NIH (NIDDK grant no. R01 DK116004) and with institutional support from the University of Florida College of Pharmacy. Research in the Reimann/Gribble laboratories was funded by the Wellcome Trust (grant nos. 106262/Z/14/Z and 106263/Z/14/Z) and the MRC (grant no. MRC_MC_UU_12012/3). Research in the Rinaman laboratory was funded by the US National Institutes of Health (grant nos. MH059911 and DK100685).

Author information

Authors and Affiliations

Authors

Contributions

The concept was developed by D.I.B., M.K.H., W.L., G.L. and S.T. The methodology was designed by D.I.B., M.K.H., F.M.G., F.R., G.L. and S.T. The formal analysis was conducted by D.I.B., A.S. and M.M. The investigation was carried out by D.I.B., M.K.H., A.S., A.A., M.M., M.V., M.H.A., S.J.L., C.M., K.S., G.L. and S.T. Resources were obtained by D.I.B., K.S., W.L., E.K., A.K., F.M.G., F.R., L.R., G.L. and S.T. Data were curated by D.I.B. The original draft was written by D.I.B., G.L. and S.T. Review and editing of the manuscript were conducted by all authors. Visualization was done by D.I.B., M.K.H. and S.T. The work was supervised by D.I.B., K.S., W.L., E.K., A.K., L.R., G.L. and S.T. Project administration was done by D.I.B., G.L. and S.T. Funding was acquired by D.I.B., W.L., F.M.G., F.R., L.R., G.L. and S.T. (contributions are defined according to the CRediT taxonomy).

Corresponding authors

Correspondence to Guillaume de Lartigue or Stefan Trapp.

Ethics declarations

Competing interests

The F.R. and F.M.G. laboratory receives funding from AstraZeneca, Eli Lilly and LGM for unrelated research and F.M.G. consults for Kallyope (New York). All other authors have no competing interests to declare.

Additional information

Peer review information Nature Metabolism thanks David D’Alessio, Kamal Rahmouni and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: George Caputa.

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

Extended data

Extended Data Fig. 1 PPGNTS neurons selectively encode large meal satiation.

a, Experimental model and paradigm for metabolic phenotyping of PPGNTS-DTA mice (DTA, n = 8) or mCherry-transduced controls (mCh, n = 7). n = 8 (DTA) / 7 (mCh) animals for analyses presented in b-o. b, Cumulative hourly food intake over 1 day, 2-way mixed-model ANOVA: Virus F(1,13)=0.015, p = 0.904. c, Daily food intake by sex, 2-way mixed-model ANOVA: Virus F(1,11)=0.012, p = 0.914; Sex F(1,11)=0.683, p = 0.426. d, Directed ambulatory locomotion (excluding fine movements) over 1 day, 2-way mixed-model ANOVA: Virus F(1,11)=0.493, p = 0.497. e-i, Meal pattern and metabolic parameters over 1 day, unpaired 2-tailed t-test or Mann-Whitney U test: (e) U = 25, p = 0.779; (f) t(13)=0.997, p = 0.337; (g) t(13)=0.565, p = 0.582; (h) t(13)=0.797, p = 0.440; (i) t(13)=0.323, p = 0.752. j, Mean bodyweight over the 24 h test period, unpaired 2-tailed t-test: t(13)=0.883, p = 0.393. k,l, Food intake during dark and light phases, unpaired 2-tailed t-test: (k) t(13)=0.668, p = 0.516; (l) t(13)=1.251, p = 0.233. m, 24 h water intake, unpaired 2-tailed t-test: t(13)=0.205, p = 0.841. n, Ensure liquid diet preload intake, unpaired 2-tailed t-test: t(8)=0.219, p = 0.832; and post-Ensure chow intake, Mann-Whitney U test: U = 2, p = 0.038. o, Post-fast refeed intake, unpaired 2-tailed t-test: t(12)=2.501, p = 0.028. p-q, Hourly and cumulative intakes over 1 day from ad libitum eating PPGNTS-hM4Di mice (n = 8 animals), 2-way within-subjects ANOVA: (p) Drug F(1,7)=0.241, p = 0.639; (q) Drug F(1,7)=0.411, p = 0.542. r, Raster plot of chow pellet retrievals during the dark phase. Plots from the same mouse after saline and CNO injections presented adjacently. All data presented as mean ± SEM.

Extended Data Fig. 2 PPGNTS neurons suppress eating without behavioural disruption.

a, Non-cumulative hourly food intake over the circadian cycle from ad libitum eating PPGNTS-hM3Dq mice (n = 7 animals for analyses presented in a-b), 2-way within-subjects ANOVA: Drug x Time F(23,138)=4.599, p < 0.0001. b, Dark phase food intake by sex, 2-way mixed-model ANOVA: Drug F(1,5)=19.97, p = 0.0066; Sex F(1,5)=3.854, p = 0.107. c,d, Photomicrographs of colocalized cFos immunoreactivity and DIO-hM3Dq-mCherry in NTS of PPG-Cre:tdRFP mice perfused 3 hours after injection of saline or CNO (photomicrographs representative of independent experiments from 4/3 animals), cc: central canal. Scale=100 µm (inset 50 µm). e, Proportion of mCherry-expressing neurons co-localised with cFos-ir in mice perfused after administration of saline or CNO (n = 4 (SAL) / 3 (CNO) animals), unpaired 1-tailed t-test: t(5)=13.94, p < 0.0001. f, Non-cumulative hourly food intake over 1 day from 18 h fasted PPGNTS-hM3Dq mice (n = 7 animals for analyses presented in f-h), 2-way within-subjects ANOVA: Drug x Time F(23,138)=3.745, p < 0.0001. The behavioural satiety sequence (BSS) was analysed during the first 40 minutes of the dark phase. g,h, Food intake and eating rate over 40 minute BSS test, paired 2-tailed t-test and Wilcoxon matched pairs test: (g) t(6)=4.088, p = 0.0064; (h) W = 18, p = 0.156. All data presented as mean ± SEM.

Extended Data Fig. 3 Glp1r-expressing VANs suppress eating and condition flavour avoidance.

a–c, Light phase food intake and metabolic parameters from ad libitum eating GLP-1RNodose-hM3Dq mice (n = 7 animals for analyses presented in a-c), paired 2-tailed t-test: (a) t(6)=0.0141, p = 0.989; (b) t(6)=0.952, p = 0.378; (c) t(6)=0.0406, p = 0.969. d, Photomicrographs of cFos immunoreactivity (cFos-ir) in coronal NTS sections from GLP-1R-Cre x PPG-YFP mice bilaterally injected in nodose ganglia with dye (Control) or AAV9-DIO-hM3Dq-mCherry (hM3Dq) and administered saline or CNO (photomicrographs representative of independent experiments from 3/3 animals). Distance in mm posterior to Bregma in bottom left, cc: central canal. Scale=100μm. e, cFos immunoreactive cells in the NTS (mean per section) of control) and hM3Dq mice (n = 3 animals per group for analyses in e-f), unpaired 2-tailed t-test: t(4)=2.981, p = 0.0407. f, PPGNTS neurons colocalised with cFos immunoreactivity in the NTS. Mann-Whitney 2-tailed U-test: U = 0, p = 0.100. g, Photomicrograph of nodose ganglion section from GLP-1R-Cre:tdRFP mouse injected with AAV encoding Cre-dependent channelrhodopsin and eYFP fluorescent reporter (DIO-CHR2-eYFP), and colocalisation of the tdRFP and eYFP reporters (photomicrographs representative of independent experiments from 4 animals). Scale=100 µm. h,i, Quantification of viral transduction specificity (h; co-localised cells as % (±SEM) of all eYFP+ cells) and efficiency (i; colocalised cells as % (±SEM) of all tdRFP+ cells), from a total of 374 tdRFP+ cells and 366 eYFP+ cells from the nodose ganglia of 3 mice. All data presented as mean ± SEM.

Extended Data Fig. 4 Oxtr rather than Glp1r VANs are the major vagal input to PPGNTS neurons.

a, Photomicrograph of coronal NTS section from PPG-Cre:tdRFP mouse transduced with DIO-TVA-mCherry + DIO-RabiesG, and subsequently with rabies virus-ΔG-GFP (RABV). Bilateral NTS injection of TVA + RabiesG and counterbalanced unilateral injection of RABV (4 mice / side) resulted in 40.5% (±5.5) of all PPGNTS neurons being successfully transduced ‘starter’ neurons, identified by colocalisation of mCherry (and/or tdRFP) and GFP (photomicrographs in a-h representative of independent experiments from 8 animals). Despite unilateral RABV injection, starter neurons were observed in left and right NTS in all mice, indicating substantial viral spread and bilateral transduction. Scale=100μm. b, Total RABV + cells in left and right nodose ganglia (LNG / RNG; n = 6 / 7 biologically independent samples), unpaired 2-tailed t-test: t(11)=0.214, p = 0.834. c,d, Quantification of Glp1r and Oxtr colocalisation in nodose ganglia (c), and proportions of dual-expressing Glp1r / Oxtr cells colocalised with RABV (d). This dual population comprises 24.7% of all Glp1r cells and 19.7% of all Oxtr cells. 9% of RABV + vagal inputs to PPGNTS neurons express both Glp1r and Oxtr, and 26.1% of dual-expressing Glp1r / Oxtr cells are RABV + vagal inputs to PPGNTS neurons. e, Quantification of RABV and Glp1r colocalisation in NG as proportions of all RABV + cells and all Glp1r+ cells, including those Glp1r cells that also express Oxtr. f, Quantification of RABV and Oxtr colocalisation in NG as proportions of all RABV + cells and all Oxtr+ cells, including those Oxtr cells that also express Glp1r. g,h, Photomicrographs of left and right nodose ganglion sections showing rabies virus GFP expression (RABV) and Glp1r and Oxtr FISH. RABV + Glp1r colocalisation shown by white arrows, RABV + Oxtr by green arrows and RABV + Glp1r+Oxtr by white-edged green arrow. Scale=100μm. All data presented as mean ± SEM.

Extended Data Fig. 5 PPGNTS neurons are necessary for oxytocin-induced eating suppression.

a,b, Food intake and bodyweight change over 1 day in eGFP and DTA mice (n = 5 (DTA) / 7 (eGFP) animals) administered oxytocin (0.4 mg/kg, i.p.), 2-way mixed-model ANOVA: (a) Drug F(1,10)=0.00474, p = 0.947; (b) Drug F(1,10)=0.0989, p = 0.760. c-d, Photomicrographs of coronal NTS sections from PPG-Cre:GCaMP3 mice injected with eGFP control virus (c) or DTA virus (d). Note the complete absence of green (GCaMP3-expressing, amplified by immunostaining against the GFP antigen) PPGNTS neurons in DTA-ablated tissue, and the extent of viral spread as demonstrated by constitutive expression of mCherry (photomicrographs representative of independent experiments from 7/5 animals). Distance in mm posterior to Bregma in bottom left, cc: central canal. Scale=100μm. All data presented as mean ± SEM.

Extended Data Fig. 6 PPGNTS neurons are not a major synaptic target of area postrema Glp1r neurons.

a, Photomicrographs of coronal NTS section showing RABV expression, Glp1r FISH and TH-ir. RABV + Glp1r+TH-ir colocalisation shown by white-edged green arrows (photomicrographs representative of independent experiments from 4 animals). Scale=100μm (inset 20μm). b,c, Quantification of Glp1r and TH-ir co-localization in area postrema (b), and proportions of dual Glp1r / TH-ir cells co-localised with RABV (c). This dual population comprises 49.4% of all TH-ir cells and 31.2% of all Glp1r cells. 9.7% of RABV + AP inputs to PPGNTS neurons express Glp1r and are TH-ir, and 2.7% of dual Glp1r / TH-ir cells are RABV + AP inputs to PPGNTS neurons. All data presented as mean ± SEM.

Extended Data Fig. 7 Liraglutide and semaglutide suppress eating independently of PPGNTS neurons.

a-e, Cumulative food intake by virus at 1,2,4,6 and 21 hr in eGFP and DTA mice (n = 8 (DTA) / 7 (eGFP) animals for analyses presented in (a-j) administered liraglutide (200 μg/kg, s.c.), 2-way mixed-model ANOVA: (a) Drug F(1,13)=0.246, p = 0.628; (b) Drug F(1,13)=2.108, p = 0.170; (c) Drug F(1,13)=37.44, p < 0.0001, Virus F(1,13)=0.836, p = 0.377; (d) Drug F(1,13)=75.09, p < 0.0001, Virus F(1,13)=1.877, p = 0.194; (e) Drug F(1,13)=154.9, p < 0.0001, Virus F(1,13)=1.272, p = 0.280. f-j, Cumulative food intake by virus at 1,2,4,6 and 21 hr in eGFP and DTA mice administered semaglutide (60 μg/kg, s.c.), 2-way mixed-model ANOVA: (f) Drug F(1,13)=1.965, p = 0.184; (g) Drug F(1,13)=17.1, p = 0.0012; Virus F(1,13)=0.630, p = 0.442; (h) Drug F(1,13)=82.49, p < 0.0001, Virus F(1,13)=0.332, p = 0.574; (i) Drug F(1,13)=98.21, p < 0.0001, Virus F(1,13)=0.840, p = 0.376; (j) Drug F(1,13)=126.1, p < 0.0001, Virus F(1,13)=3.42, p = 0.0873. k-m, Representative photomicrographs of cFos immunoreactivity (cFos-ir) in arcuate nucleus of the hypothalamus (ARC) 4 hours after vehicle (VEH, n = 4 animals) or semaglutide (SEMA, 60 μg/kg, s.c., n = 4 animals) administration, and total cFos count, unpaired 1-tailed t-test: m) t(6)=2.614, p = 0.020. Scale=100μm. (n-p) Representative photomicrographs of cFos-ir in paraventricular nucleus of the hypothalamus (PVN) 4 hours after vehicle or semaglutide administration (n = 4 / 4 animals), and total cFos count, unpaired 1-tailed t-test: (p) t(6)=5.109, p = 0.0011. Scale=100μm. q-t, Representative photomicrographs of cFos-ir in dorsal lateral and external lateral subdivisions of the parabrachial nucleus (dlPBN / elPBN) 4 hours after vehicle or semaglutide administration (n = 3 / 4 animals), and total cFos count, unpaired 1-tailed t-tests: (s) t(5)=1.693, p = 0.0756; (t) t(5)=3.57, p = 0.0080. Semaglutide did not increase cFos-ir in the medial PBN, t(5)=0.435, p = 0.341. Scale=100μm. All data presented as mean ± SEM.

Extended Data Fig. 8 PPGNTS neuron activation augments semaglutide-induced eating suppression.

a-d, Bodyweight change at 24 and 48 hours, and cumulative food intake at 48 and 72 hours (n = 6 animals), 1-way within-subjects ANOVA: (a) Drug F(2.1,10.5)=61.61, p < 0.0001; (b) Drug F(2.3,11.3)=102.7, p < 0.0001; (c) Drug F(2.1,10.6)=24.38, p < 0.0001; (d) Drug F(1.9,9.3)=40.35, p < 0.0001. 72 hr BW data not shown: Drug F(2.0,10.2)=4.22, p = 0.0454, no significant pairwise comparisons. e, Photomicrographs of coronal NTS sections from PPG-Cre:GCaMP3 mice injected with AAV encoding Cre-dependent hM3Dq and mCherry fluorescent reporter (DIO-hM3Dq-mCherry), and colocalisation of the GCaMP3 (amplified by immunostaining against GFP antigen) and mCherry reporters (photomicrographs representative of independent experiments from 4 animals). Distance in mm from Bregma in bottom left, cc: central canal. Scale=100 µm. f,g, Quantification of viral transduction specificity (f; co-localised cells as % (±SEM) of all mCherry+ cells) and efficiency (g; co-localised cells as % (±SEM) of all GCaMP3+ cells), from a total of 410 mCherry+ cells and 391 GCaMP3+ cells from 4 mice. All data presented as mean ± SEM.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brierley, D.I., Holt, M.K., Singh, A. et al. Central and peripheral GLP-1 systems independently suppress eating. Nat Metab 3, 258–273 (2021). https://doi.org/10.1038/s42255-021-00344-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00344-4

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