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:

High-fat food biases hypothalamic and mesolimbic expression of consummatory drives

Abstract

Maintaining healthy body weight is increasingly difficult in our obesogenic environment. Dieting efforts are often overpowered by the internal drive to consume energy-dense foods. Although the selection of calorically rich substrates over healthier options is identifiable across species, the mechanisms behind this choice remain poorly understood. Using a passive devaluation paradigm, we found that exposure to high-fat diet (HFD) suppresses the intake of nutritionally balanced standard chow diet (SD) irrespective of age, sex, body mass accrual and functional leptin or melanocortin-4 receptor signaling. Longitudinal recordings revealed that this SD devaluation and subsequent shift toward HFD consumption is encoded at the level of hypothalamic agouti-related peptide neurons and mesolimbic dopamine signaling. Prior HFD consumption vastly diminished the capacity of SD to alleviate the negative valence associated with hunger and the rewarding properties of food discovery even after periods of HFD abstinence. These data reveal a neural basis behind the hardships of dieting.

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: HFD exposure promotes weight gain and devaluation of SD.
Fig. 2: HFD-exposure alters ARCAgRP activity responses to SD and HFD in fasted mice.
Fig. 3: HFD exposure induces alterations in DMHLepR responses to SD.
Fig. 4: HFD-exposure disrupts ARCAgRP responses to both peripheral detection of calories and signals of hunger.
Fig. 5: HFD-exposure disrupts functional capacity of ARCAgRP neurons to drive SD, but not HFD, consumption.
Fig. 6: HFD alters ARCNpy-mediated VTADat responses to SD.
Fig. 7: HFD exposure disrupts functional capacity of VTADat neurons to potentiate SD consumption.
Fig. 8: HFD exposure dampens AcbSh DA release in response to SD.

Similar content being viewed by others

Data availability

Data and supporting materials will be made available by the corresponding authors upon reasonable request.

Code availability

Code is available by the corresponding authors upon request or directly at https://www.niehs.nih.gov/research/atniehs/labs/ln/pi/iv/tools/index.cfm.

References

  1. Timper, K. & Brüning, J. C. Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis. Model. Mech. 10, 679–689 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. DiFeliceantonio, A. G. & Small, D. M. Dopamine and diet-induced obesity. Nat. Neurosci. 22, 1–2 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Ferrario, C. R. et al. Homeostasis meets motivation in the battle to control food intake. J. Neurosci. 36, 11469–11481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Luquet, S., Perez, F. A., Hnasko, T. S. & Palmiter, R. D. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310, 683–685 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Aponte, Y., Atasoy, D. & Sternson, S. M. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat. Neurosci. 14, 351–355 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. 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 

  7. Takahashi, K. A. & Cone, R. D. Fasting induces a large, leptin-dependent increase in the intrinsic action potential frequency of orexigenic arcuate nucleus neuropeptide y/agouti-related protein neurons. Endocrinology 146, 1043–1047 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Mandelblat-Cerf, Y. et al. Arcuate hypothalamic AgRP and putative pomc neurons show opposite changes in spiking across multiple timescales. eLife 4, 1–25 (2015).

    Article  Google Scholar 

  9. Chen, Y., Lin, Y.-C., Kuo, T.-W. & Knight, Z. A. Sensory detection of food rapidly modulates arcuate feeding circuits. Cell 160, 829–841 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Beutler, L. R. et al. Dynamics of gut–brain communication underlying hunger. Neuron 96, 461–475 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Su, Z., Alhadeff, A. L. & Betley, J. N. Nutritive, post-ingestive signals are the primary regulators of AgRP neuron activity. Cell Rep. 21, 2724–2736 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Betley, J. N. et al. Neurons for hunger and thirst transmit a negative-valence teaching signal. Nature 521, 180–185 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Baver, S. B. et al. Leptin modulates the intrinsic excitability of AgRP/NPY neurons in the arcuate nucleus of the hypothalamus. J. Neurosci. 34, 5486–5496 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Salamone, J. D., Correa, M., Mingote, S. & Weber, S. M. Nucleus accumbens dopamine and the regulation of effort in food-seeking behavior: implications for studies of natural motivation, psychiatry, and drug abuse. J. Pharmacol. Exp. Ther. 305, 1–8 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Berridge, K. C. ‘Liking’ and ‘wanting’ food rewards: brain substrates and roles in eating disorders. Physiol. Behav. 97, 537–550 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wise, R. A. Role of brain dopamine in food reward and reinforcement. Philos. Trans. R. Soc. B Biol. Sci. 361, 1149–1158 (2006).

    Article  CAS  Google Scholar 

  17. Alhadeff, A. L. et al. Natural and drug rewards engage distinct pathways that converge on coordinated hypothalamic and reward circuits. Neuron 103, 891–908.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Denis, R. G. P. et al. Palatability can drive feeding independent of AgRP neurons. Cell Metab. 22, 646–657 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fordahl, S. C. & Jones, S. R. High-fat-diet-induced deficits in dopamine terminal function are reversed by restoring insulin signaling. ACS Chem. Neurosci. 8, 290–299 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Rothemund, Y. et al. Differential activation of the dorsal striatum by high-calorie visual food stimuli in obese individuals. Neuroimage 37, 410–421 (2007).

    Article  PubMed  Google Scholar 

  21. Stice, E., Spoor, S., Bohon, C., Veldhuizen, M. G. & Small, D. M. Relation of reward from food intake and anticipated food intake to obesity: a functional magnetic resonance imaging study. J. Abnorm. Psychol. 117, 924–935 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Thanarajah, S. E. et al. Food intake recruits orosensory and post-ingestive dopaminergic circuits to affect eating desire in humans. Cell Metab. 29, 695–706 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Ravussin, Y. et al. Effects of chronic weight perturbation on energy homeostasis and brain structure in mice. Am. J. Physiol. Integr. Comp. Physiol. 300, R1352–R1362 (2011).

    Article  CAS  Google Scholar 

  24. Johnson, P. M. & Kenny, P. J. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat. Neurosci. 13, 635–641 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cone, J. J., Chartoff, E. H., Potter, D. N., Ebner, S. R. & Roitman, M. F. Prolonged high fat diet reduces dopamine reuptake without altering DAT gene expression. PLoS ONE 8, e58251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Drewnowski, A. & Greenwood, M. R. C. Cream and sugar: human preferences for high-fat foods. Physiol. Behav. 30, 629–633 (1983).

    Article  CAS  PubMed  Google Scholar 

  27. Yang, Y. Jr, D. L, S., Keating, K. D., Allison, D. B. & Nagy, T. R. Variations in body weight, food intake and body composition after long-term high-fat diet feeding in C57BL/6J mice. Obesity 22, 2147–2155 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Guo, J., Jou, W., Gavrilova, O. & Hall, K. D. Persistent diet-induced obesity in male C57BL/6 mice resulting from temporary obesigenic diets. PLoS ONE 4, e5370 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Carlin, J. L. et al. Removal of high-fat diet after chronic exposure drives binge behavior and dopaminergic dysregulation in female mice. Neuroscience 326, 170–179 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Balthasar, N. et al. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 123, 493–505 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Jais, A. & Brüning, J. C. Hypothalamic inflammation in obesity and metabolic disease. J. Clin. Invest. 127, 24–32 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Chen, T.-W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Garfield, A. S. et al. Dynamic GABAergic afferent modulation of AgRP neurons. Nat. Neurosci. 19, 1628–1635 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hahn, T. M., Breininger, J. F., Baskin, D. G. & Schwartz, M. W. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat. Neurosci. 1, 271–272 (1998).

    Article  CAS  PubMed  Google Scholar 

  36. Briggs, D. I., Enriori, P. J., Lemus, M. B., Cowley, M. A. & Andrews, Z. B. Diet-induced obesity causes ghrelin resistance in arcuate NPY/AgRP neurons. Endocrinology 151, 4745–4755 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Briggs, D. I. et al. Calorie-restricted weight loss reverses high-fat diet-induced ghrelin resistance, which contributes to rebound weight gain in a ghrelin-dependent manner. Endocrinology 154, 709–717 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, S. et al. Consumption of palatable food primes food approach behavior by rapidly increasing synaptic density in the VTA. Proc. Natl Acad. Sci. USA 113, 2520–2525 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Roitman, M. F., Stuber, G. D., Phillips, P. E. M., Wightman, R. M. & Carelli, R. M. Dopamine operates as a subsecond modulator of food seeking. J. Neurosci. 24, 1265–1271 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Atasoy, D., Aponte, Y., Su, H. H. & Sternson, S. M. A FLEX switch targets channelrhodopsin-2 to multiple cell types for imaging and long-range circuit mapping. J. Neurosci. 28, 7025–7030 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S. & Roth, B. L. Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc. Natl Acad. Sci. USA 104, 5163–5168 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Alexander, G. M. et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron 63, 27–39 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tsai, H.-C. et al. Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science 324, 1080–1084 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sun, F. et al. A genetically encoded fluorescent sensor enables rapid and specific detection of dopamine in flies, fish, and mice. Cell 174, 481–496 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Betley, J. N., Cao, Z. F. H., Ritola, K. D. & Sternson, S. M. Parallel, redundant circuit organization for homeostatic control of feeding behavior. Cell 155, 1337–1350 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lippert, R. N. et al. Maternal high-fat diet during lactation reprograms the dopaminergic circuitry in mice. J. Clin. Invest. 130, 3761–3776 (2020).

  47. Tellez, L. A. et al. Separate circuitries encode the hedonic and nutritional values of sugar. Nat. Neurosci. 19, 465–470 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Stice, E., Yokum, S., Blum, K. & Bohon, C. Weight gain is associated with reduced striatal response to palatable food. J. Neurosci. 30, 13105–13109 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Volkow, N. D., Wang, G.-J. & Baler, R. D. Reward, dopamine and the control of food intake: implications for obesity. Trends Cogn. Sci. 15, 37–46 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Meng, C. et al. Spectrally resolved fiber photometry for multi-component analysis of brain circuits. Neuron 98, 707–717 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

Download references

Acknowledgements

Research was supported by an NIEHS–NIDDK joint fellowship. We thank N. Martin and B. Gloss of the NIEHS Viral Vector Core for producing AAVs and J. Tucker of the NIEHS Fluorescence Microscopy and Imaging Center for assistance with image acquisition. We thank the GENIE project for the development of GCaMP6. We would also like to thank all members of Dr. Krashes’ and Dr. Cui’s labs for their technical support and guidance throughout this work and J. Cushman of the NIEHS Neurobehavioral Core for assistance with behavioral studies and statistical analyses. This work was supported by the Intramural Research Program of the National Institutes of Health, the National Institute of Environmental Health Sciences (1ZIAES103310 to G.C.), the National Institutes of Diabetes and Digestive and Kidney Diseases (DK075088 to M.J.K. and DK075087-06 to M.J.K.), the NIEHS–NIDDK Joint Fellowship Award (to C.M.M.) and the Center of Compulsive Behaviors (to C.M.M. and I.D.A.S.).

Author information

Authors and Affiliations

Authors

Contributions

C.M.M., J. L.-G. and M.J.K. designed experiments with technical input from G.C., J. L.-G., N.S.W. and M.H.B. M.S. performed and analyzed home cage consumption and body composition, fast–refeed and optogenetic experiments. C.M.M. performed and analyzed home cage AgRP fiber photometry experiments and DA sensor experiments. C.L. performed and analyzed LepR-Cre fiber photometry experiments. C.L. and I.D.A.S. performed electrophysiology recordings. J. L.-G. performed and analyzed gastric infusion and VTA studies. F.S., Y.Z. and Y.L. provided the DA2m sensor and provided technical guidance. N.P.K. and D.M.R. performed histological verification and imaging. C.M.M., J. L.-G. and M.J.K. wrote the manuscript with input from G.C., C.L., I.D.A.S., N.S.W., M.H.B., M.S., N.P.K., D.M.R., F.S, Y.Z. and Y.L.

Corresponding authors

Correspondence to Guohong Cui or Michael J. Krashes.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Paul Kenny and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 HFD-exposure promotes fat mass accrual, reduction of homecage SD intake and devaluation of SD in physiologically hungry mice independent of weight gain.

Weekly a, fat mass (n = 20, males and females, RM two-way ANOVA, Week x Group: F (11, 418) = 50.17, P < 0.0001, Sidak’s multiple comparisons test) and b, lean mass throughout diet exposure (n = 20, males and females, RM two-way ANOVA, Week x Group: F (11, 418) = 0.8075, P = 0.6326). Daily c, body weights and d, SD intake throughout the duration of the experiment of individual animals exposed to HFD. e, No correlation between the average amount of SD intake during the HFD-exposure period and body weight changes after 8 weeks of SD and HFD access (n = 20, males and females, Linear regression, R2 = 0.02713, P = 0.4877). f, Between-subject comparison of 1 hr SD fast-refeed consumption across test sessions (n = 16 SD group, n = 18 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (4, 128) = 28.99, P < 0.0001, Bonferroni’s multiple comparisons test). g, No correlation between SD devaluation at Week 8 relative to Baseline and body weight changes after 8 weeks of access to SD and HFD (n = 18, males and females, Linear regression, R2=0.001090, P = 0.8965). h, Experimental timeline, group schematic for home cage measurements and i, and daily body weights throughout the duration of the experiment (n = 12 per group, males and females). j, Within-subject comparison of 1 hr SD fast-refeed consumption across testing sessions (n = 12 limited HFD weight-matched to SD group, males and females, RM one-way ANOVA, Time: F (2.431, 26.74) = 66.81, P < 0.0001, Tukey’s multiple comparisons). k, Within-subject, within-diet comparisons of 1 hr SD and HFD fast-refeed consumption across testing sessions (n = 18, males and females, RM one-way ANOVAs, Tukey’s multiple comparisons). Dotted lines in a–d and i delineate window of HFD availability. All error bars and shaded areas in a, b and i represent mean ± s.e.m. Shaded blue area in f, j, and k represent HFD homecage availability. *P < 0.05, **P < 0.01, ****P < 0.0001.

Extended Data Fig. 2 Intact melanocortin-4 receptor- or leptin-signaling are dispensable for SD devaluation following HFD-exposure and 24 h of HFD-exposure is sufficient to devalue SD.

a, d Experimental timeline and group schematic for fast-refeed test with 1 hr SD access. b, e Within-subject and c, f between-subject comparisons of 1 hr SD fast-refeed consumption across testing sessions (n = 5 per Mc4r KO + /- and -/- group, males and females, RM two-way ANOVA, Time x Group: F (4, 32) = 0.3173, P = 0.8643, Tukey’s multiple comparisons) (n = 4 per Leptin KO + /- and -/- group, males and females, RM two-way ANOVA, Time x Group: F (4, 24) = 0.3313, P = 0.8542, Tukey’s multiple comparisons). g, Experimental timeline and group schematic for fast-refeed test with 1 hr SD access. h, Within-subject and i, between-subject comparisons of 1 hr SD fast-refeed consumption across testing sessions (n = 16 per group, males and females, RM two-way ANOVA, Time x Group: F (1, 30) = 16.96, P = 0.0003, Sidak’s multiple comparisons test). B = Baseline. WD = withdrawal. Shaded blue area in b, c, e, f, h and i represent HFD homecage availability. All error bars represent s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 3 Acute and chronic HFD-exposure alters ARCAgRP activity responses to SD and HFD in fasted mice largely independent of body-weight changes.

a, Between-subject quantification of fasted ARCAgRP activity response to Pellet 1 (SD) (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 16.74, P < 0.0001, Bonferroni’s multiple comparisons) and b, SD consumed across testing sessions (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 15.81, P < 0.0001, Bonferroni’s multiple comparisons). c–f, ARCAgRP activity changes to Pellet 1 (SD) presentation are not strongly correlated with bodyweight accrual over the entire length of the experiment (n = 12 SD group, n = 11 SD + HFD group, males and females, Linear regression, c, SD group: R2 = 0.001058, P = 0.9201, SD + HFD group: R2 = 0.6731, P = 0.0020, d, SD group: R2 = 0.2304, P = 0.1142, SD + HFD group: R2 = 0.1154, P = 0.3067, e, SD group: R2 = 0.1539, P = 0.2072, SD + HFD group: R2 = 0.1319, P = 0.2723, f, SD group: R2 = 0.002643, P = 0.8739, SD + HFD group: R2 = 0.2958, P = 0.0837) g, Between-subject quantification of fasted ARCAgRP activity response to Pellet 2 (SD for SD group; SD for SD + HFD group during B1 and B2 and HFD for SD + HFD group during 1, 4, 8 and 10 weeks) (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 21.07, P < 0.0001, Bonferroni’s multiple comparisons) and h, total calories consumed across testing sessions (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 14.26, P < 0.0001, Bonferroni’s multiple comparisons). i, Experimental timeline and group schematic for fasted photometry recordings. j, Average fasted photometry traces across recording sessions aligned to Pellet 1 and 2 presentation (n = 6, males and females). k, Within-subject quantification of fasted ARCAgRP activity response to Pellet 1 (SD) (n = 6, males and females, Paired t test (two-tailed), P = 0.0099) and l, SD consumed across testing sessions (n = 6, males and females, Paired t test (two-tailed), P = 0.0049). m, Within-subject quantification of fasted ARCAgRP activity response to Pellet 2 (SD for Baseline; HFD for 1 day) (n = 6, males and females, Paired t test (two-tailed), P = 0.0008) and n, total calories consumed across testing sessions (n = 6, males and females, Paired t test (two-tailed), P = 0.0008). Shaded blue area in a, b, and g, h represent HFD homecage availability. B1 and B2 refer to Baseline 1 and 2, respectively. WD = withdrawal. Dotted lines in j indicate Pellet 1 and Pellet 2 presentation. All error bars and shaded regions of j represent s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 4 HFD-exposure alters ARCAgRP activity responses to HFD in fed mice.

a, Experimental timeline and group schematic for fed photometry recordings. b, c, Average fed photometry traces of the b, SD group and c, SD + HFD group across recording sessions aligned to Pellet 1 and 2 presentation (n = 12 SD group, n = 11 SD + HFD group, males and females). d, Within- and e, between-subject quantification of fed ARCAgRP activity response to Pellet 1 (SD) (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 0.8953, P = 0.4872, Bonferroni’s multiple comparisons). f, Within- and g, between-subject quantification of SD consumed across testing sessions (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 1.768, P = 0.1258). h, Within- and i, between-subject quantification of fed ARCAgRP activity response to Pellet 2 (SD for SD group; SD for SD + HFD group during B1 and B2 and HFD for SD + HFD group during 1, 4, 8 and 10 weeks) (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 7.565, P < 0.0001, Bonferroni’s multiple comparisons). j, Within- and k, between-subject quantification of total calories consumed across testing sessions (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 31.68, P < 0.0001, Bonferroni’s multiple comparisons). Dotted lines in b, c indicate Pellet 1 and Pellet 2 presentation. Shaded blue area in d–k represent HFD homecage availability. B1 and B2 refer to Baseline 1 and 2, respectively. WD = withdrawal. All error bars and shaded regions of b, c represent s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 5 Spectrum-based fiber photometry reveals diminished ARCAgRP baseline activity following HFD exposure.

a, Representative GCaMP and tdTomato emission spectra from a mouse during the Fed and Fasted sessions of Baseline 1 prior to the addition of food. b, GCaMP/tdTomato ratios normalized to the mean of Baseline 1 + 2 Fed values shows basal ARCAgRP activity is increased during Fasted sessions relative to Fed sessions (SD – Fed vs Fasted; HFD – Fed vs Fasted). Homecage HFD exposure reduces basal ARCAgRP activity relative to the SD group across both Fed and Fasted conditions (Fed – SD vs HFD; Fasted – SD vs HFD) (n = 12 SD group, n = 11 SD + HFD group, males and females, three-way ANOVA, Diet: F(1,21) = 6.515, P = 0.019, Satiety state: F(1,63) = 94.128, P < 0.0001) c, Fasted GCaMP/tdTomato ratios normalized to the Fed values across recording sessions demonstrates an ~50% fasting-induced increase in the GCaMP/tdTomato ratio across all recording weeks (n = 12 SD group, n = 11 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (5, 105) = 1.978, P = 0.0879). B1 and B2 refer to Baseline 1 and 2, respectively. WD = withdrawal.

Extended Data Fig. 6 Physiological disruption of ARCAgRP activity in response to HFD-exposure is partially dependent on body weight gain.

a, Changes in ARCAgRP activity in response to identical caloric load is correlated to weight gain 1, but not 4 weeks after HFD exposure or 1 week HFD withdrawal (n = 6, males and females, Linear regression, 1 week (HFD): R2 = 0.6903, P = 0.0405, 4 weeks (HFD): R2 = 0.1356, P = 0.4727, 5 weeks (WD): R2 = 0.003755, P = 0.9082). b-e, Changes in ARCAgRP activity in response to ghrelin is correlated to weight gain (n = 12 SD group, n = 11 SD + HFD group, males and females, Linear regression, b, SD group: R2 = 0.01466, P = 0.7078, SD + HFD group: R2 = 0.3905, P = 0.0398, c, SD group: R2 = 0.05629, P = 0.4578, SD + HFD group: R2 = 0.4724, P = 0.0194, d, SD group: SD group: R2 = 0.1504, P = 0.2129, SD + HFD group: R2 = 0.2336, P = 0.1321), e, SD group: R2 = 0.2907, P = 0.0705, SD + HFD group: R2 = 0.4042, P = 0.0355. f–k, Changes in ARCAgRP activity in response to f, g, CCK, h–I, 5HT or j, k, PYY is not correlated to weight gain (n = 5 SD group, n = 6 SD + HFD group, males and females, Linear regression, f, SD group: R2 = 0.008391, P = 0.8835, SD + HFD group: R2 = 0.09392, P = 0.5547, g, SD group: R2 = 0.003959, P = 0.9199, SD + HFD group: R2 = 0.06259, P = 0.6326, h, SD group: R2 = 0.6037, P = 0.1221, SD + HFD group: R2 = 0.0009157, P = 0.9546, i, SD group: R2 = 0.6415, P = 0.1034, SD + HFD group: R2 = 0.0007729, P = 0.9583, j, SD group: R2 = 0.3514, P = 0.2922, SD + HFD group: R2 = 0.05138, P = 0.6602, k, SD group: R2 = 0.09218, P = 0.6194, SD + HFD group: R2 = 0.01706, P = 0.8052). l, n, p, Average photometry traces of the SD group across recording sessions aligned to l, CCK, n, 5HT or p, PYY injection (n = 5 SD group, males and females). m, o, q, Within-subject quantification of ARCAgRP activity to m, CCK (n = 5 SD group, males and females, RM two-way ANOVA, Time Bin x Week: F (1.511, 6.045) = 1.014, P = 0.3923), o, 5HT (n = 5 SD group, males and females, RM two-way ANOVA, Time Bin x Week: F (2.334, 9.337) = 1.242, P = 0.3393) or q, PYY (n = 5 SD group, males and females, RM two-way ANOVA, Time Bin x Week: F (1.897, 7.589) = 1.885, P = 0.2164) across testing sessions. Dotted lines indicate l, CCK, n, 5HT or p, PYY injection. B1 and B2 refer to Baseline 1 and 2, respectively. All error bars and shaded regions of l, n, and p represent s.e.m.

Extended Data Fig. 7 HFD exposure promotes devaluation of optogenetic ARCAgRP-evoked SD, but not HFD, and consumption independent of weight gain.

a, Between-subject comparison of fedAgRP stimulation 1 hr SD consumption across test sessions (n = 16 SD group, n = 17 SD + HFD group, males and females, RM two-way ANOVA, Time x Group: F (4, 124) = 18.85, P < 0.0001, Bonferroni’s multiple comparisons). b, No correlation between SD devaluation at Week 8 relative to Baseline and body weight changes after 8 weeks of access to SD and HFD (n = 17, males and females, Linear regression, R2 = 0.04964, P = 0.3900). c, Within-subject, within-diet comparisons of fedAgRP stimulation 1 hr SD and HFD consumption across testing sessions (n = 17, males and females, RM one-way ANOVAs, Tukey’s multiple comparisons). B = Baseline. WD = Withdrawal. Shaded blue area in a and c represent HFD homecage availability. All error bars represent s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 8 ARCAgRP/Npy activation does not alter basal VTADat activity but potentiates signaling in response to food via an indirect signaling mechanism.

a, Brain schematic of anterograde tracing strategy. b–d, representative images of AgRP::Synaptophysin-tdTomato and tyrosine hydroxylase (TH) in the b, arcuate nucleus, c, paraventricular hypothalamus and d, ventral tegmental area. e, Brain schematic of unilateral viral delivery of Cre-inducible ChR2 to the ARC of AgRP-ires-Cre mice and position of ex vivo slice recordings from PVH (top) and VTA (bottom) cells. f, g, ChR2-assisted circuit mapping schematic and representative trace from f, PVH and g, VTA cells. h, Representative image of Dat-ires-Cre::Ai32-ChR2-eYFP and overlap with TH in the ARC. i, Chemogenetic ARCNpy activation drives food intake (n = 6, males and females, Paired t test (two-tailed), P = 0.0013). j, Averaged Z-score traces and k, quantification of basal VTADat activity after saline or CNO injections (n = 6, males and females, Paired t test (two-tailed), P = 0.2837). l, Averaged Z-score traces and m, mean (n = 6, males and females, Paired t test (two-tailed), P = 0.0656) and n, max quantification (n = 6, males and females, Paired t test (two-tailed), P = 0.0365) of VTADat activity in response to SD food drop after saline or CNO injections. All error bars represent s.e.m. *P < 0.05, **P < 0.01.

Extended Data Fig. 9 VTADat photostimulation is intrinsically rewarding but loses the capacity to enhance SD consumption in fasted mice after HFD exposure.

a, Schematic of Real-Time Place Preference assay. b, Averaged heat maps and quantification of time spent in each chamber of the RTPP assay in VTADat::ChR2 mice (n = 9, males, Paired t test (two-tailed), P < 0.0001). c, Averaged heat maps and quantification of time spent in each chamber of the RTPP assay in control VTADat::YFP mice (n = 6, males, Paired t test (two-tailed), P = 0.9576). d, Within-subject and condition comparisons of 1 hr SD consumption across testing sessions with or without VTADat photostimulation (n = 9, males, RM two-way ANOVA, Time x Condition: F (3, 48) = 18.30, P < 0.0001, Tukey’s multiple comparisons). B = Baseline. WD = Withdrawal. Shaded blue area in d represents HFD homecage availability. All error bars represent s.e.m. ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 10 DA responses to SD are state-dependent and consistent over time in HFD-naïve animals.

a, Averaged Z-score traces of DA2m fluorescence aligned to SD pellet retrieval across appetite state (n = 8, males and females). b, Within-subject comparisons of mean fluorescence Z-score during SD pellet retrieval across appetite state (n = 8, males and females, Paired t test (two-tailed), P = 0.0659). c, Within-subject comparisons of SD pellets consumed across appetite state (n = 8, males and females, Paired t test (two-tailed), P < 0.0001). d, Averaged Z-score traces of DA2m fluorescence aligned to SD pellet retrieval across recording weeks. (n = 4, males and females) e, Within-subject comparisons of mean fluorescence Z-score during SD pellet retrieval across sessions (n = 4, males and females, Wilcoxon matched-pairs signed rank test, P = 0.625). f, Within-subject comparisons of SD pellets consumed across sessions (n = 4, males and females, Wilcoxon matched-pairs signed rank test, P > 0.9999). Time 0 denotes consumption. B = Baseline. Shaded peach region in a and d represent quantified pellet approach/retrieval period. All error bars and shaded regions of a and d represent s.e.m. ****P < 0.0001.

Supplementary information

Reporting Summary

Supplementary Table 1

Supplementary Table 1. Statistics.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mazzone, C.M., Liang-Guallpa, J., Li, C. et al. High-fat food biases hypothalamic and mesolimbic expression of consummatory drives. Nat Neurosci 23, 1253–1266 (2020). https://doi.org/10.1038/s41593-020-0684-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-020-0684-9

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