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:

Dissociable hindbrain GLP1R circuits for satiety and aversion

Abstract

The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.

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: Hindbrain GLP1R neurons mediate the anorexic and weight-loss effects of GLP1-based obesity drugs.
Fig. 2: In vivo activity dynamics of APGLP1R and NTSGLP1R neurons.
Fig. 3: APGLP1R and NTSGLP1R neurons drive aversion and satiety, respectively.
Fig. 4: Obesity drugs reduce food intake when aversion circuitry (APGLP1R neurons) is blunted.
Fig. 5: APGLP1R and NTSGLP1R neurons form discrete projections to mediate functionally dissociable behaviours.

Similar content being viewed by others

Data availability

Source data are provided with this paper. Transcriptomic analyses were generated from publicly-accessible data (NCBI GEO databases: GSE160938, GSE166649, GSE228192). Source data are provided with this paper.

Code availability

Custom codes generated to analyse data from the study are accessible at https://github.com/alhadefflab/2p_imaging_analysis.git.

References

  1. Wadden, T. A. et al. Effect of subcutaneous semaglutide vs placebo as an adjunct to intensive behavioral therapy on body weight in adults with overweight or obesity: The STEP 3 Randomized Clinical Trial. JAMA 325, 1403–1413 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Wilding, J. P. H. et al. Once-weekly semaglutide in adults with overweight or obesity. N. Engl. J. Med. 384, 989–1002 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. World Obesity Federation. World Obesity Atlas 2023 (World Obesity Federation, 2023).

  4. Jastreboff, A. M. et al. Triple-hormone-receptor agonist retatrutide for obesity — a phase 2 trial. N. Engl. J. Med. 389, 514–526 (2023).

    Article  CAS  PubMed  Google Scholar 

  5. Jastreboff, A. M. et al. Tirzepatide once Wweekly for the treatment of obesity. N. Engl. J. Med. 387, 205–216 (2022).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hayes, M. R., De Jonghe, B. C. & Kanoski, S. E. Role of the glucagon-like-peptide-1 receptor in the control of energy balance. Physiol. Behav. 100, 503–510 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. Gabery, S. et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight 5, https://doi.org/10.1172/jci.insight.133429 (2020).

  11. Jensen, C. B. et al. Characterization of the glucagonlike peptide-1 receptor in male mouse brain using a novel antibody and in situ hybridization. Endocrinology 159, 665–675 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. Zhang, C. et al. Area postrema cell types that mediate nausea-associated behaviors. Neuron 109, 461–472 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Xie, Z. et al. The gut-to-brain axis for toxin-induced defensive responses. Cell 185, 4298–4316 (2022).

    Article  CAS  PubMed  Google Scholar 

  15. Grill, H. J. & Hayes, M. R. Hindbrain neurons as an essential hub in the neuroanatomically distributed control of energy balance. Cell Metab. 16, 296–309 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ludwig, M. Q. et al. A genetic map of the mouse dorsal vagal complex and its role in obesity. Nat. Metab. 3, 530–545 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Adriaenssens, A. et al. Hypothalamic and brainstem glucose-dependent insulinotropic polypeptide receptor neurons employ distinct mechanisms to affect feeding. 8, https://doi.org/10.1172/jci.insight.164921 (2023).

  18. Lindberg, J. S. et al. Cinacalcet HCl, an oral calcimimetic agent for the treatment of secondary hyperparathyroidism in hemodialysis and peritoneal dialysis: a randomized, double-blind, multicenter study. J. Am. Soc. Nephrol. 16, 800–807 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Harrington, P. E. & Fotsch, C. Calcium sensing receptor activators: calcimimetics. Curr. Med. Chem. 14, 3027–3034 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Kanoski, S. E., Rupprecht, L. E., Fortin, S. M., De Jonghe, B. C. & Hayes, M. R. The role of nausea in food intake and body weight suppression by peripheral GLP-1 receptor agonists, exendin-4 and liraglutide. Neuropharmacology 62, 1916–1927 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Pelchat, M. L., Grill, H. J., Rozin, P. & Jacobs, J. Quality of acquired responses to tastes by Rattus norvegicus depends on type of associated discomfort. J. Comp. Psychol. 97, 140–153 (1983).

    Article  CAS  PubMed  Google Scholar 

  22. Grill, H. J. & Norgren, R. The taste reactivity test. I. Mimetic responses to gustatory stimuli in neurologically normal rats. Brain Res. 143, 263–279 (1978).

    Article  CAS  PubMed  Google Scholar 

  23. Watakabe, A. et al. Comparative analyses of adeno-associated viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neurosci. Res. 93, 144–157 (2015).

    Article  PubMed  Google Scholar 

  24. Wilson-Pérez, H. E. et al. Vertical sleeve gastrectomy is effective in two genetic mouse models of glucagon-like Peptide 1 receptor deficiency. Diabetes 62, 2380–2385 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Murphy, S. et al. Nucleus of the solitary tract A2 neurons control feeding behaviors via projections to the paraventricular hypothalamus. Neuropsychopharmacology 48, 351–361 (2023).

    Article  CAS  PubMed  Google Scholar 

  26. Rinaman, L. Ascending projections from the caudal visceral nucleus of the solitary tract to brain regions involved in food intake and energy expenditure. Brain Res. 1350, 18–34 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, D. et al. Whole-brain mapping of the direct inputs and axonal projections of POMC and AgRP neurons. Front. Neuroanat. 9, 40 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Carter, M. E., Han, S. & Palmiter, R. D. Parabrachial calcitonin gene-related peptide neurons mediate conditioned taste aversion. J. Neurosci. 35, 4582–4586 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Campos, C. A., Bowen, A. J., Schwartz, M. W. & Palmiter, R. D. Parabrachial CGRP neurons control meal termination. Cell Metab 23, 811–820 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Carter, M. E., Soden, M. E., Zweifel, L. S. & Palmiter, R. D. Genetic identification of a neural circuit that suppresses appetite. Nature 503, 111–114 (2013).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  31. Li, M. M. et al. The paraventricular hypothalamus regulates satiety and prevents obesity via two genetically distinct circuits. Neuron 102, 653–667 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, S. X. et al. Competition between stochastic neuropeptide signals calibrates the rate of satiation. Preprint at bioRxiv https://doi.org/10.1101/2023.07.11.548551 (2023).

  33. Fenselau, H. et al. A rapidly acting glutamatergic ARC→PVH satiety circuit postsynaptically regulated by α-MSH. Nat. Neurosci. 20, 42–51 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Spector, A. C., Breslin, P. & Grill, H. J. Taste reactivity as a dependent measure of the rapid formation of conditioned taste aversion: a tool for the neural analysis of taste-visceral associations. Behav. Neurosci. 102, 942–952 (1988).

    Article  CAS  PubMed  Google Scholar 

  35. Dowsett, G. K. C. et al. A survey of the mouse hindbrain in the fed and fasted states using single-nucleus RNA sequencing. Mol Metab 53, 101240 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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 

  37. Göke, R., Larsen, P. J., Mikkelsen, J. D. & Sheikh, S. P. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur. J. Neurosci. 7, 2294–2300 (1995).

    Article  PubMed  Google Scholar 

  38. Alhadeff, A. L. & Grill, H. J. Hindbrain nucleus tractus solitarius glucagon-like peptide-1 receptor signaling reduces appetitive and motivational aspects of feeding. Am. J. Physiol. Regul. Integr. Comp. Physiol. 307, R465–R470 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fortin, S. M. 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 

  40. Hayes, M. R. et al. Intracellular signals mediating the food intake-suppressive effects of hindbrain glucagon-like peptide-1 receptor activation. Cell Metab. 13, 320–330 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Alhadeff, A. L. et al. Endogenous glucagon-like peptide-1 receptor signaling in the nucleus tractus solitarius is required for food intake control. Neuropsychopharmacology 42, 1471–1479 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Costa, A. et al. Anorectic and aversive effects of GLP-1 receptor agonism are mediated by brainstem cholecystokinin neurons, and modulated by GIP receptor activation. Mol. Metab. 55, 101407 (2022).

    Article  CAS  PubMed  Google Scholar 

  43. Ilanges, A. et al. Brainstem ADCYAP1+ neurons control multiple aspects of sickness behaviour. Nature 609, 761–771 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  44. 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 

  45. Burmeister, M. A. et al. The hypothalamic glucagon-like peptide 1 receptor Is sufficient but not necessary for the regulation of energy balance and glucose homeostasis in mice. Diabetes 66, 372–384 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Hayes, M. R., Skibicka, K. P. & Grill, H. J. Caudal brainstem processing is sufficient for behavioral, sympathetic, and parasympathetic responses driven by peripheral and hindbrain glucagon-like-peptide-1 receptor stimulation. Endocrinology 149, 4059–4068 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. D’Agostino, G. et al. Nucleus of the solitary tract serotonin 5-HT2C receptors modulate food intake. Cell Metab. 28, 619–630 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Webster, A. N. et al. Molecular connectomics reveals a glucagon-like peptide 1 sensitive neural circuit for satiety. Preprint at bioRxiv https://doi.org/10.1101/2023.10.31.564990 (2023).

  49. D’Agostino, G. et al. Appetite controlled by a cholecystokinin nucleus of the solitary tract to hypothalamus neurocircuit., eLife 5, e12225 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Roman, C. W., Sloat, S. R. & Palmiter, R. D. A tale of two circuits: CCKNTS neuron stimulation controls appetite and induces opposing motivational states by projections to distinct brain regions. Neuroscience 358, 316–324 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Cheng, W. et al. Calcitonin receptor neurons in the mouse nucleus tractus solitarius control energy balance via the non-aversive suppression of feeding. Cell Metab. 31, 301–312 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ran, C., Boettcher, J. C., Kaye, J. A., Gallori, C. E. & Liberles, S. D. A brainstem map for visceral sensations. Nature 609, 320–326 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  53. Ly, T. et al. Sequential appetite suppression by oral and visceral feedback to the brainstem. Nature 624, 130–137 (2023).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  54. Shu, Y. et al. Gastrointestinal adverse events associated with semaglutide: a pharmacovigilance study based on FDA adverse event reporting system. Front. Public Health 10, 996179 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Atasoy, D., Betley, J. N., Su, H. H. & Sternson, S. M. Deconstruction of a neural circuit for hunger. Nature 488, 172–177 (2012).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  56. Wu, Q., Boyle, M. P. & Palmiter, R. D. Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell 137, 1225–1234 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Egecioglu, E. et al. The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology 38, 1259–1270 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Tuesta, L. M. et al. GLP-1 acts on habenular avoidance circuits to control nicotine intake. Nat. Neurosci. 20, 708–716 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Schmidt, H. D. et al. Glucagon-like peptide-1 receptor activation in the ventral tegmental area decreases the reinforcing efficacy of cocaine. Neuropsychopharmacology 41, 1917–1928 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wickham, R. J. Nausea and vomiting: a palliative care imperative. Curr. Oncol. Rep. 22, 1 (2020).

    Article  PubMed  Google Scholar 

  61. Williams, E. 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 

  62. Madisen, L. et al. A toolbox of Cre-dependent optogenetic transgenic mice for light-induced activation and silencing. Nat. Neurosci. 15, 793–802 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Huang, K.-P. et al. Sex differences in response to short-term high fat diet in mice. Physiol. Behav. 221, 112894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Alhadeff, A. L. et al. A neural circuit for the suppression of pain by a competing need state. Cell 173, 140–152 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Goldstein, N. et al. Hypothalamic detection of macronutrients via multiple gut-brain pathways. Cell Metab. 33, 676–687 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Min, S. et al. Arterial baroreceptors sense blood pressure through decorated aortic claws. Cell Rep. 29, 2192–2201 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ghidewon, M. et al. Growth differentiation factor 15 (GDF15) and semaglutide inhibit food intake and body weight through largely distinct, additive mechanisms. Diabetes Obes. Metab. 24, 1010–1020 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wyvell, C. L. & Berridge, K. C. Intra-accumbens amphetamine increases the conditioned incentive salience of sucrose reward: enhancement of reward “wanting” without enhanced “liking” or response reinforcement. J. Neurosci. 20, 8122–8130 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Berridge, K. C. & Peciña, S. Benzodiazepines, appetite, and taste palatability. Neurosci. Biobehav. Rev. 19, 121–131 (1995).

    Article  CAS  PubMed  Google Scholar 

  70. Ren, D. et al. A prokaryotic voltage-gated sodium channel. Science 294, 2372–2375 (2001).

    Article  CAS  PubMed  ADS  Google Scholar 

  71. Giovannucci, A. et al. CaImAn an open source tool for scalable calcium imaging data analysis., eLife 8, e38173 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Pnevmatikakis, E. A. & Giovannucci, A. NoRMCorre: an online algorithm for piecewise rigid motion correction of calcium imaging data. J. Neurosci. Methods 291, 83–94 (2017).

    Article  CAS  PubMed  Google Scholar 

  73. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  74. Pnevmatikakis, E. A. et al. Simultaneous denoising, deconvolution, and demixing of calcium imaging data. Neuron 89, 285–299 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tan, H.-E. et al. The gut–brain axis mediates sugar preference. Nature 580, 511–516 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  76. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ludwig, M. Q., Todorov, P. V., Egerod, K. L., Olson, D. P. & Pers, T. H. Single-cell mapping of GLP-1 and GIP receptor expression in the dorsal vagal complex. Diabetes 70, 1945–1955 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Dietrich and H. Grill for comments on the manuscript; B. Arenkiel and J. Ortiz for providing viral reagents; R. Seeley for providing transgenic mice; P. Bazzino for advice on intraoral catheters; H. Grill for advice on taste-reactivity experiments; C. Ran for advice on two-photon imaging experiments; and Monell Center facilities and administrative staff for animal care and other experimental support. A.L.A. is a New York Stem Cell Foundation Robertson investigator and a Pew biomedical scholar. This work was supported by the National Institutes of Health (R00DK119574 and DP2AT011965 to A.L.A.); the American Heart Association (857082 to A.L.A. and 898990 to K.-P.H.); the New York Stem Cell Foundation (to A.L.A.); the Klingenstein Fund and Simons Foundation (to A.L.A.); the Pew Charitable Trusts (to A.L.A.); the National Science Foundation (2236662 to M.Y.G.); the Penn Institute for Diabetes, Obesity, and Metabolism (to A.L.A.); and the Monell Chemical Senses Center (to K.A.B. and A.L.A.). The confocal microscope used in these studies was purchased with instrumentation grant NIH S10OD030354 (to A.L.A.).

Author information

Authors and Affiliations

Authors

Contributions

K.-P.H., M.Y.G., A.D.M. and A.L.A. conceived and designed the experiments. K.-P.H., A.A.A., M.Y.G., A.D.M., M.S.A., N.T.N., N.D.H., N.P., Y.S.K.G., A.E.A., K.A.B. and A.L.A. performed experiments, analysed data and/or interpreted data; A.L.A. wrote the manuscript with comments from K.-P.H. and all authors.

Corresponding author

Correspondence to Amber L. Alhadeff.

Ethics declarations

Competing interests

The authors declare that the Monell Chemical Senses Center has filed a patent application related to potential therapeutic compounds.

Peer review

Peer review information

Nature thanks Mark Andermann, Chuchu Zhang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 DVCGLP1R neurons are required for the food intake suppression and weight loss effects of GLP1-based obesity drugs.

a, Representative image and quantification of viral expression (EGFP) in Glp1r-expessing neurons of Glp1r-ires-Cre mice (RNA in situ hybridization). b, Representative images of Glp1r expression (RNA in situ hybridization) in the DVC, ARC, and NG of control mice or mice with GLP1R neuron ablation in each region. c, Quantification of Caspase3- or DTA-mediated neural ablation: relative number (to controls) of GLP1R+ neurons in DVC (left), ARC (middle), and NG (right) of mice with viral injection into DVC, ARC, or NG, respectively (n = 3-5 mice/group, two-sided unpaired t-tests, all ps < 0.01). d, Food intake following IP injection of saline or exendin-4 in fasted control or GLP1R-ablated mice (n = 8-14 mice/group, two-way repeated measures ANOVAs, all ps < 0.01 except DVC where p=ns). e, Body weight at 8 weeks post-surgery in control, DVC, ARC, and NG GLP1R neuron ablation groups (n = 7-8 mice/group, one-way ANOVA, p=ns). f, Percent body weight change during the onset of diet-induced obesity in high-fat diet-fed control (EGFP) or DVCGLP1R neuron-ablated mice receiving semaglutide (n = 11-15 mice/group, two-way repeated measures ANOVA, control versus DVC-Casp3, p < 0.001). gi, Fat mass (g, n = 11-15 mice/group, two-way ANOVA, Control versus DVC-Casp at 8 and 12 weeks: p < 0.001), lean mass (h, n = 11-15/group, two-way ANOVA, all ps=ns) and cumulative food intake (i, n = 11-13 mice/group, two-sided unpaired t-test, p < 0.05) in high-fat diet-fed control or DVC GLP1R-ablated mice over 12 weeks of biweekly SQ semaglutide. Values are mean ± S.E.M. ¤¤p < 0.01, ¤¤¤p < 0.001 drug x time interaction; *p < 0.05, **p < 0.01, ***p < 0.001 post hoc comparisons. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 2 DVCGLP1R neuron activation suppresses food intake and energy expenditure.

a, Schematic for continuous food intake and energy expenditure measurements using mouse metabolic chambers. b, Cumulative food intake in high-fat diet-fed control mice or mice with chronic NaChBac-mediated DVCGLP1R neuron activation (n = 7-8 mice/group, two-way repeated measures ANOVA, control versus NaChBac: p < 0.001). Shaded areas represent dark periods. c, Average inter-meal interval of high-fat diet-fed control mice or mice with chronic NaChBac-mediated DVCGLP1R neuron activation (n = 7-8 mice/group, two-sided unpaired t-test, control versus NaChBac: p < 0.01). d, Average meal size in high-fat diet-fed control mice or mice with chronic NaChBac-mediated DVCGLP1R neuron activation (n = 7-8 mice/group, two-sided unpaired t-test, control vs. NaChBac: p=ns). e, Energy expenditure in high-fat diet-fed control mice or mice with chronic NaChBac-mediated DVCGLP1R neuron activation (n = 7-8 mice/group). Shaded area represents dark period. f, Dark period energy expenditure normalized to lean mass by ANCOVA in high-fat diet-fed control mice or mice with chronic NaChBac-mediated DVCGLP1R neuron activation (n = 7-8 mice/group, ANCOVA, p = 0.06). g, Energy expenditure in fasted chow-fed control mice or mice with chemogenetic DVCGLP1R neuron activation (n = 8 mice/group). Shaded area represents dark period. h, Dark period energy expenditure normalized to body mass by ANCOVA in chow-fed control mice or mice with chemogenetic DVCGLP1R neuron activation (n = 8 mice/group, ANCOVA, p < 0.001). Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 3 Imaging APGLP1R and NTSGLP1R neurons.

a, Coronal images of soma-restricted AAV5-EF1a-DIO-EGFPL10a in the DVC of Glp1r-ires-Cre mice across the rostral-caudal axis. Scale bar, 100 µm. b, Quantification of GLP1R neurons in AP and NTS across the rostral-caudal axis (n = 3 mice/group). c, Coronal images of soma-restricted AAV5-EF1a-DIO-EGFPL10a in the DVC of Glp1r-ires-Cre;Ai9(tdTomato) mice. Images depict the dense GLP1R projection into the NTS but not into the AP. Scale bar, 100 µm. d, Maximum projection two-photon images (transverse plane) of the DVC from Glp1r-ires-Cre;Ai9(tdTomato) mice injected with Cre-dependent GCaMP6s depicting the red halo of GLP1R projections into the NTS, part of the strategy to draw boundaries between AP and NTS (see Methods for additional details). Scale bar, 100 µm. e, Two-photon images of the DVC from Glp1r-ires-Cre;Ai9(tdTomato) mice injected with Cre-dependent GCaMP6s across the z-axis. AP and NTS neurons are visible on all imaging planes because of the angle of the medulla for imaging (see Methods for additional details). f, Top, schematic for simultaneous in vivo two-photon imaging of APGLP1R and NTSGLP1R neurons in mice. Bottom, images of surgical prep and cranial window for in vivo two-photon imaging of calcium dynamics in APGLP1R and NTSGLP1R neurons. g, Proportion of APGLP1R and NTSGLP1R neurons activated by semaglutide (n = 5 mice, two-sided paired t-test, p=ns). h, Heat maps depicting z-scored responses of semaglutide-activated APGLP1R (n = 678 neurons, 5 mice) and NTSGLP1R (n = 388 neurons, 5 mice) neurons sorted by activation time. i, Median time to semaglutide-induced activation in APGLP1R and NTSGLP1R neurons depicted in h (n = 5 mice, two-sided unpaired t-test, p=ns). j, Mean time to semaglutide-induced activation in APGLP1R and NTSGLP1R neurons depicted in h (n = 5 mice, two-sided unpaired t-test, p=ns). Values are mean ± S.E.M. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 4 In vivo responses of APGLP1R and NTSGLP1R neurons to nutritive and aversive stimuli.

a, Heat maps depicting z-scored responses of APGLP1R neurons (n = 375 neurons, 5 mice) and NTSGLP1R neurons (n = 266 neurons, 5 mice) to saline or Ensure. Dashed white lines indicate start of stimulus. b, Average z-scored activity responses in APGLP1R neurons (blue) and NTSGLP1R neurons (green) to saline or Ensure administration (n = 6 mice, two-way ANOVA, AP: saline versus Ensure: p < 0.01, NTS: saline versus Ensure p < 0.001). c, Individual data points comparing APGLP1R (blue) and NTSGLP1R (green) neuron z-scored activity responses to saline and semaglutide. Dotted lines represent threshold for statistically significant neural activation (z = 1.64, see Methods for additional details). d, Individual heat maps for each mouse (n = 7) depicting z-scored responses of APGLP1R and NTSGLP1R neurons to Ensure or cinacalcet. e, Representative two-photon images of GCaMP6s across the z axis with neurons colour-coded based on responses to Ensure (most responsive in blue, index = −10) and cinacalcet (most responsive in red, index=10). AP/NTS boundary indicated in yellow. f, Individual heat maps for each mouse (n = 6) depicting z-scored responses of APGLP1R and NTSGLP1R neurons to Ensure or LiCl. g, Probability calculations for activation of individual cells by Ensure or aversive stimuli (n = 6-7 mice/group, two-sided paired t-tests, p < 0.001). We compare the product of the probabilities of a neuron being activated by either Ensure or aversive stimuli [P(Ensure) x P(cinacalcet) or P(Ensure) x P(LiCl)] to the probability of a neuron being activated by both stimuli [P(Ensure and cinacalcet) or P(Ensure and LiCl)]. That the probability of a cell being activated by both stimuli is generally lower than the product of individual probabilities suggests a level of specialization of individual cells where most are biased toward nutritive or aversive stimuli, with less overlap than would be expected by chance. Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 5 Comparison of motion correction strategies for extracting calcium dynamics.

To determine whether alternate motion correction strategies improved calcium imaging analyses in our setup, we compared rigid-body motion correction (“Rigid”) with two other strategies (and the two strategies combined). First, we applied the piecewise rigid function (“pw-rigid”) in NoRMCorre to create a grid within each of our images, where each patch of the grid was corrected to better adjust for non-rigid distortions. Second, we applied PyStackReg to realign all frames using an affine transformation. We compared the resulting proportions of activated neurons and individual neuron calcium traces for each method. Although these alternate motion correction strategies did not significantly change our data, we note for readers that some imaging preparations (especially those that have more ROIs around the edges of frames) may require such strategies. a, Comparison of proportions of neurons activated by nutritive (Ensure, blue), aversive (cinacalcet, pink), or both (green) stimuli in APGLP1R and NTSGLP1R neurons across one z-level from each of two example mice chosen at random, after imaging analysis following different motion correction strategies. b, Comparison of individual neuron calcium traces in APGLP1R and NTSGLP1R neurons activated by nutritive (Ensure, blue), aversive (cinacalcet, pink), or both (green) stimuli after imaging analysis following different motion correction strategies. Traces were chosen pseudo-randomly, excluding neurons that were not selected as ROIs in each of the four analyses [these, however, were included in a, where we did not exclude any neurons], and accounting for representation of Ensure-activated, cinacalcet-activated, and both-activated neurons where possible.

Source Data

Extended Data Fig. 6 Real-time and conditioned orofacial taste reactivity in mice.

a,b, Schematics of stereotyped hedonic (a) and aversive (b) orofacial responses in mice. Modified from schematics in ref. 22. c,d, Composite score (c, n = 5-10 mice/group, one-way ANOVA, all ps<0.05) and individual behaviours (d, n = 5-10 mice/group, two-way ANOVA, all ps<0.001) for hedonic taste reactivity responses to quinine concentrations (0, 2.4, 4 mM). e,f, Composite score (e, n = 5-10 mice/group, one-way ANOVA, all ps<0.01) and individual behaviours (f, 5-10 mice/group, two-way ANOVA, all ps<0.05) for aversive taste reactivity responses to quinine concentrations (0, 2.4, 4 mM). g,h, Composite scores (g, n = 6 mice/group, two-sided paired t-tests, ps<0.05) and individual behaviours (h, 6 mice/group, two-way ANOVA, RMM: p < 0.05, G: p < 0.001) for real-time hedonic and aversive taste reactivity responses to a flavour paired with cinacalcet (15 µmol/kg, IP). i,j, Composite scores (i, n = 6 mice/group, two-sided paired t-tests, aversive: p < 0.05) and individual behaviours (j, 6 mice/group, two-way ANOVA, G: p < 0.001) for conditioned hedonic and aversive taste reactivity responses to a flavour paired with cinacalcet (15 µmol/kg, IP). k,l, Composite scores (k, n = 5 mice/group, two-sided paired t-tests, ps<0.01) and individual behaviours (l, 6 mice/group, two-way ANOVA, RMM: p < 0.001, PL: p < 0.05, G: p < 0.001, CR: p < 0.001) for real-time hedonic and aversive taste reactivity responses to a flavour paired with LiCl (6 mmol/kg, IP). m,n, Composite scores (m, n = 5 mice/group, two-sided paired t-tests, ps<0.05) and individual behaviours (n = 6 mice/group, two-way ANOVA, RMM: p < 0.05, G: p < 0.001, CR: p < 0.05) for conditioned hedonic and aversive taste reactivity responses to a flavour paired with LiCl (6 mmol/kg, IP). o, Schematic for chemogenetic stimulation of DVCGLP1R neurons. p,q, Individual hedonic taste reactivity behaviours at baseline and after conditioning in control (p, n = 6 mice/group, two-way ANOVA, all ps=ns) and experimental (q, n = 7 mice/group, two-way ANOVA, RMM: p < 0.001, LTP: p < 0.05) mice. r,s, Individual aversive taste reactivity behaviours at baseline and after conditioning in control (r, n = 6 mice/group, two-way ANOVA, all ps=ns) and experimental (s, n = 7 mice/group, two-way ANOVA, G: p < 0.01, CR: p < 0.001) mice. Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 7 Number of APGLP1R, but not NTSGLP1R, neurons expressing chemogenetic receptors correlates with avoidance behaviour.

a, Representative image of hM3Dq expression in the DVC (AP and NTS). b, Correlation between the number of hM3Dq-expressing neurons (x-axis) and behaviour [y-axis; avoidance (CS+ intake in CFA assay) or anorexia (food intake) elicited by DVCGLP1R neuron stimulation] in the AP (left graphs) or NTS (right graphs) (n = 9 mice, Pearson correlation, p < 0.05 for AP/avoidance, other ps=ns). Red colour indicates statistical significance. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 8 APGLP1R and NTSGLP1R neurons are largely unique populations of hindbrain neurons with dissociable behavioural effects.

a,b, Schematic and representative images of viral (AAV2.2-FLEX-tdTomato) separation of AP (a) and NTS (b) using 20 nl intracranial injections. c, Quantification of tdTomato-expressing GLP1R neurons in AP and NTS after viral injection into AP (n = 3 mice). d, Quantification of tdTomato-expressing GLP1R neurons in AP and NTS after viral injection into NTS (n = 3 mice). e, Strategy for viral injections to directly compare behavioural results of activation of DVCGLP1R, APGLP1R, and NTSGLP1R neurons: we used the same viral serotype and injection protocol to activate neurons in each of these regions. f, 12-h food intake in mice following activation of DVCGLP1R, APGLP1R, and NTSGLP1R neurons (n = 7-11 mice/group, two-way ANOVA, all ps<0.001). g, Meal size in fasted mice following chemogenetic activation of APGLP1R or NTSGLP1R neurons (n = 10-11 mice/group, two-way ANOVA, APGLP1R-hM3Dq vehicle versus CNO: p < 0.001, NTSGLP1R-hM3Dq vehicle versus CNO: p=ns). h,i, Energy expenditure in fasted mice with chemogenetic APGLP1R (h) or NTSGLP1R (i) neuron activation (n = 11 mice/group). Shaded area represents dark period. j, Dark period energy expenditure normalized to body mass by ANCOVA in mice with chemogenetic APGLP1R (blue) or NTSGLP1R (green) neuron activation (n = 11 mice/group, ANCOVA, p=ns). k, Schematic describing protocol for taste reactivity experiments. l,m, Individual hedonic taste reactivity behaviours at baseline and after conditioning with chemogenetic APGLP1R (l, n = 8 mice, two-way ANOVA, RMM baseline versus conditioned p < 0.001) or NTSGLP1R (m, n = 5 mice, two-way ANOVA, all ps=ns) neuron activation. n,o, Individual aversive taste reactivity behaviours at baseline and after conditioning with chemogenetic APGLP1R (n, n = 8 mice, two-way ANOVA, CR baseline versus conditioned: p < 0.001) or NTSGLP1R (o, n = 5 mice, two-way ANOVA, all ps=ns) neuron activation. p, Uniform manifold approximation and projection (UMAP) plot showing AP and NTS neuron subtypes. Analysis was made after combining data sets from ref. 13, ref. 16, and ref. 17, which all contained cells from both the AP and NTS. q, Dot plots indicating normalized expression of genes in Glp1r+ cells of the AP and NTS. Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 9 Deletion of GLP1R in DVC attenuates anorexia and aversion by obesity drugs.

a, Schematic for deletion of GLP1R in DVC neurons using Glp1rfl/fl mice and injection of AAV2.2-Cre. b, Quantification of efficacy of GLP1R deletion: relative number of Glp1r+ neurons in the DVC in WT and Glp1rfl/fl mice after DVC injection of AAV2.2-Cre (n = 3 mice/group, two-sided unpaired t-test, p < 0.001). c, Mice underwent a conditioned flavour avoidance assay to semaglutide. d, Intake of flavour paired with semaglutide (CS+) in control (WT) or DVC GLP1R-deleted (Glp1rfl/fl) mice before (baseline) and after conditioning (n = 6-8 mice/group, two-way ANOVA, WT baseline versus conditioned: p < 0.001, Glp1rfl/fl baseline vs. conditioned: p=ns). e, Food intake (4 h) was measured in response to exendin-4 or semaglutide. For exendin-4 studies, food was returned immediately after injection. For semaglutide studies, food was returned 4 h post-injection. f, Food intake in response to exendin-4 in WT and Glp1rfl/fl mice (e, n = 6-7 mice/group, two-way ANOVA, WT: p < 0.01, Glp1rfl/fl: p=ns). g, Food intake in response to semaglutide in WT and Glp1rfl/fl mice (e, n = 7-8 mice/group, two-way ANOVA, WT: p < 0.05, Glp1rfl/fl: p=ns). Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Extended Data Fig. 10 Representative images and injection verifications for neural tracing experiments.

a, Representative image of viral expression of AAV5-DIO-ChR2-EYFP at the injection site (DVC: AP and NTS) of Glp1r-ires-Cre mice (n = 3 mice). cc, central canal; DMV, dorsal motor nucleus of the vagus. bf, Representative images of DVCGLP1R axons in brain regions involved in feeding behaviour: lPBN (b), PVH (c), ARC (d), BNST (e), CeA (f). Boxes indicate zoomed in regions. 3 V, third ventricle; ac, anterior commissure; BNST, bed nucleus of the stria terminalis; CeA, central amygdala; LV, lateral ventricle; Scp, superior cerebellar peduncle. g, Left, Cre-dependent H129-ΔTK-TT was injected into the DVC of Glp1r-ires-Cre mice (n = 3). Right, representative image of starter cells for anterograde H129-ΔTK-TT-mediated tracing from DVCGLP1R neurons. h, Schematic of mechanism for fluorophore expression using PRV-263. i, Glp1r-ires-Cre mice (n = 3 mice/group) were injected in the lPBN or PVH with PRV-263 and DVC brain sections were imaged for retrogradely-transported Cre-positive (GLP1R+, mCerulean/EYFP, green) and Cre-negative (GLP1R−, tdTomato, red) neurons in the AP and NTS. j, Representative image of lPBN injection site. Scale bar, 200 µm. k, Representative image of PVH injection site. Scale bar, 100 µm. l, Representative images of PRV-263 in Cre-positive (GLP1R+, mCerulean/EYFP, green) and Cre-negative (GLP1R-, tdTomato, red) neurons of the AP and NTS after viral injection in the lPBN (left) or PVH (right). Distances indicate mm from bregma. Scale bar, 50 µm. m, Representative image of injection site in AP after injection of Cre-dependent AAV2.2-DIO-ChR2-EYFP in APGLP1R neurons in Glp1r-ires-Cre mice (n = 3 mice). Scale bar, 50 µm. n, Representative image of PVH in mouse with EYFP injection in APGLP1R neurons. Scale bar, 50 µm. o, Representative images of PBN projections across the rostral-caudal axis, distances indicate mm from bregma. p, Representative image of injection site in NTS after injection of Cre-dependent AAV2.2-DIO-ChR2-EYFP in NTSGLP1R neurons in Glp1r-ires-Cre mice (n = 3 mice). Scale bar, 50 µm. q, Representative image of lPBN in mouse with EYFP injection in NTSGLP1R neurons. Scale bar, 50 µm. r, Representative images of PVH projections across the rostral-caudal axis, distances indicate mm from bregma. Scale bar, 100 µm.

Extended Data Fig. 11 Neural activation in the lPBN and PVH with DVCGLP1R neuron stimulation.

a, Schematic for RNA in situ hybridization for Fos and CGRP (Calca) in lPBN or Fos and Mc4r in PVH after chemogenetic stimulation of DVCGLP1R neurons. b, Representative images (left) and quantification (right) of colocalization of Fos and Calca in the lPBN after control (vehicle) or chemogenetic (CNO) stimulation (n = 3 mice/group, two-sided unpaired t-test, p < 0.05). elPBN, external lateral PBN; clPBN, central lateral PBN. c, Representative images (left) and quantification of colocalization (right) of Fos and Mc4r in the PVH after control (vehicle) or chemogenetic (CNO) stimulation (n = 3 mice/group, two-sided unpaired t-test, p < 0.01). Values are mean ± S.E.M. *p < 0.05, **p < 0.01.

Source Data

Extended Data Fig. 12 Behavioural effects of APGLP1R→lPBN and NTSGLP1R→PVH neuron optogenetic activation.

a,b, Food intake (1 h) in control mice expressing EYFP in APGLP1R→lPBN (a, n = 6 mice, paired t-test, p=ns) and NTSGLP1R→PVH (b, n = 5 mice, two-sided paired t-test, p=ns) neurons. (Grey bars (−), no stimulation; coloured bars (+), stimulation). c,d, Time spent in stimulation zone during real-time place avoidance assay in ad libitum-fed control mice expressing EYFP in APGLP1R→lPBN (c, n = 5-6 mice/group, two-way ANOVA, all ps=ns) or NTSGLP1R→PVH (d, n = 5 mice, two-way ANOVA, all ps=ns) neurons. e,f, Time spent in stimulation zone during real-time place avoidance assay in fasted control mice expressing EYFP in APGLP1R→lPBN (e, n = 5-6 mice/group, two-way ANOVA, all ps=ns) or NTSGLP1R→PVH (f, n = 5-6 mice/group, two-way ANOVA, all ps=ns). g,h, Time spent in stimulation zone during real-time place avoidance assay in ad libitum-fed mice expressing ChR2 in APGLP1R→lPBN (g, n = 10-11 mice/group, two-way ANOVA, Trials 2&3 no stim versus stim: ps<0.05) or NTSGLP1R→PVH (h, n = 7 mice, two-way ANOVA, all ps=ns). i,j, Time spent in stimulation zone during real-time place avoidance assay in fasted mice expressing ChR2 in APGLP1R→lPBN (i, n = 11 mice, two-way ANOVA, all ps=ns) or NTSGLP1R→PVH (j, n = 7 mice, two-way ANOVA, all ps=ns) neurons. k,l, Individual hedonic taste reactivity behaviours at baseline (pre-stimulation) and during optogenetic stimulation of APGLP1R→lPBN (k, n = 6 mice, two-way ANOVA, RMM and LTP no stim versus stim: ps<0.05) or NTSGLP1R→PVH (l, n = 6 mice, two-way ANOVA, all ps=ns) neurons. m,n, Individual aversive taste reactivity behaviours at baseline (pre-stimulation) and during optogenetic stimulation of APGLP1R→lPBN (m, n = 6 mice, two-way ANOVA, CR no stim versus stim: p < 0.001) or NTSGLP1R→PVH (n, n = 6 mice, two-way ANOVA, all ps=ns) neurons. Grey bars, no stimulation; coloured bars, optogenetic stimulation. Values are mean ± S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001. See Supplementary Table 1 for statistical details.

Source Data

Supplementary information

Supplementary Table 1

Sample sizes and statistical details for all figures and Extended Data figures.

Reporting Summary

Supplementary Video 1

3D reconstruction of DVCGLP1R neurons following injection of Glp1r-ires-Cre mouse with a Cre-dependent AAV expressing a soma-restricted fluorophore.

Supplementary Video 2

Raw, motion-corrected video (left) and dF/F video (right) of in vivo two-photon calcium imaging of DVCGLP1R neurons.

Supplementary Video 3

Examples of hedonic and aversive orofacial taste reactivity responses.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, KP., Acosta, A.A., Ghidewon, M.Y. et al. Dissociable hindbrain GLP1R circuits for satiety and aversion. Nature (2024). https://doi.org/10.1038/s41586-024-07685-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07685-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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