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:

Food odor perception promotes systemic lipid utilization

Abstract

Food cues during fasting elicit Pavlovian conditioning to adapt for anticipated food intake. However, whether the olfactory system is involved in metabolic adaptations remains elusive. Here we show that food-odor perception promotes lipid metabolism in male mice. During fasting, food-odor stimulation is sufficient to increase serum free fatty acids via adipose tissue lipolysis in an olfactory-memory-dependent manner, which is mediated by the central melanocortin and sympathetic nervous systems. Additionally, stimulation with a food odor prior to refeeding leads to enhanced whole-body lipid utilization, which is associated with increased sensitivity of the central agouti-related peptide system, reduced sympathetic activity and peripheral tissue-specific metabolic alterations, such as an increase in gastrointestinal lipid absorption and hepatic cholesterol turnover. Finally, we show that intermittent fasting coupled with food-odor stimulation improves glycemic control and prevents insulin resistance in diet-induced obese mice. Thus, olfactory regulation is required for maintaining metabolic homeostasis in environments with either an energy deficit or energy surplus, which could be considered as part of dietary interventions against metabolic disorders.

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: Fasted mice explored foods on the basis of olfactory, but not visual, information.
Fig. 2: Olfactory perception of a food cue elicits lipid mobilization during fasting.
Fig. 3: Food-odor memory recall elicits lipid mobilization during fasting.
Fig. 4: Pharmacological profiles of serum NEFA mobilization by food-odor stimulation during fasting.
Fig. 5: Acute effects of food-odor stimulation on protein and gene expression in the peripheral tissues in the fasting state.
Fig. 6: Food-odor prestimulation promotes lipid absorption and utilization upon nutrient intake.
Fig. 7: Food-odor stimulation during fasting induces tissue-specific metabolic regulations for adapting to refeeding.
Fig. 8: Effects of food-odor stimulation on metabolic functions in diet-induced obesity.

Similar content being viewed by others

Data availability

RNA-seq datasets have been deposited in the DDBJ BioProject database with the BioProject accession number PRJDB14370 (PSUB018445). All other data that support the findings of this study are presented in figures or extended data figures in this paper. Source data are provided with this paper.

References

  1. Palouzier-Paulignan, B. et al. Olfaction under metabolic influences. Chem. Senses 37, 769–797 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Julliard, A. K., Al Koborssy, D., Fadool, D. A. & Palouzier-Paulignan, B. Nutrient sensing: another chemosensitivity of the olfactory system. Front. Physiol. 8, 468 (2017).

    PubMed  PubMed Central  Google Scholar 

  3. Domjan, M. Pavlovian conditioning: a functional perspective. Annu. Rev. Psychol. 56, 179–206 (2005).

    PubMed  Google Scholar 

  4. Lasschuijt, M. P., Mars, M., de Graaf, C. & Smeets, P. A. M. Endocrine cephalic phase responses to food cues: a systematic review. Adv. Nutr. 11, 1364–1383 (2020).

    PubMed  PubMed Central  Google Scholar 

  5. Lushchak, O. V., Carlsson, M. A. & Nässel, D. R. Food odors trigger an endocrine response that affects food ingestion and metabolism. Cell. Mol. Life Sci. 72, 3143–3155 (2015).

    CAS  PubMed  Google Scholar 

  6. Mutlu, A. S., Gao, S. M., Zhang, H. & Wang, M. C. Olfactory specificity regulates lipid metabolism through neuroendocrine signaling in Caenorhabditis elegans. Nat. Commun. 11, 1450 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Jovanovic, P. & Riera, C. E. Olfactory system and energy metabolism: a two-way street. Trends Endocrinol. Metab. 33, 281–291 (2022).

    CAS  PubMed  Google Scholar 

  8. Brandt, C. et al. Food perception primes hepatic ER homeostasis via melanocortin-dependent control of mTOR activation. Cell 175, 1321–1335 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Murata, K. Hypothetical roles of the olfactory tubercle in odor-guided eating behavior. Front. Neural Circuits 14, 577880 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Kobayakawa, K. et al. Innate versus learned odour processing in the mouse olfactory bulb. Nature 450, 503–508 (2007).

    CAS  PubMed  Google Scholar 

  11. Fendt, M., Endres, T., Lowry, C. A., Apfelbach, R. & McGregor, I. S. TMT-induced autonomic and behavioral changes and the neural basis of its processing. Neurosci. Biobehav. Rev. 29, 1145–1156 (2005).

    CAS  PubMed  Google Scholar 

  12. Shirasu, M. et al. Olfactory receptor and neural pathway responsible for highly selective sensing of musk odors. Neuron 81, 165–178 (2014).

    CAS  PubMed  Google Scholar 

  13. Pérez-Gómez, A. et al. Innate predator odor aversion driven by parallel olfactory subsystems that converge in the ventromedial hypothalamus. Curr. Biol. 25, 1340–1346 (2015).

    PubMed  PubMed Central  Google Scholar 

  14. Choi, G. B. et al. Driving opposing behaviors with ensembles of piriform neurons. Cell 146, 1004–1015 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. 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).

    PubMed  PubMed Central  Google Scholar 

  16. Yang, D., Liu, T. & Williams, K. W. Motivation to eat—AgRP neurons and homeostatic need. Cell Metab. 22, 62–63 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Manceau, R., Majeur, D. & Alquier, T. Neuronal control of peripheral nutrient partitioning. Diabetologia 63, 673–682 (2020).

    PubMed  Google Scholar 

  18. Cansell, C., Denis, R. G., Joly-Amado, A., Castel, J. & Luquet, S. Arcuate AgRP neurons and the regulation of energy balance. Front. Endocrinol. 3, 169 (2012).

    CAS  Google Scholar 

  19. Kim, S. J., Windon, M. J. & Lin, S. Y. The association between diabetes and olfactory impairment in adults: a systematic review and meta-analysis. Laryngoscope Investig. Otolaryngol. 4, 465–475 (2019).

    PubMed  PubMed Central  Google Scholar 

  20. Peng, M., Coutts, D., Wang, T. & Cakmak, Y. O. Systematic review of olfactory shifts related to obesity. Obes. Rev. 20, 325–338 (2019).

    PubMed  Google Scholar 

  21. Zhang, Z. et al. Olfactory dysfunction mediates adiposity in cognitive impairment of type 2 diabetes: insights from clinical and functional neuroimaging studies. Diabetes Care 42, 1274–1283 (2019).

    CAS  PubMed  Google Scholar 

  22. Han, P., Roitzsch, C., Horstmann, A., Pössel, M. & Hummel, T. Increased brain reward responsivity to food-related odors in obesity. Obesity 29, 1138–1145 (2021).

    CAS  PubMed  Google Scholar 

  23. Fadool, D. A. et al. Kv1.3 channel gene-targeted deletion produces “super-smeller mice” with altered glomeruli, interacting scaffolding proteins, and biophysics. Neuron 41, 389–404 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Tucker, K., Overton, J. M. & Fadool, D. A. Diet-induced obesity resistance of Kv1.3–/– mice is olfactory bulb dependent. J. Neuroendocrinol. 24, 1087–1095 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Schwartz, A. B. et al. Olfactory bulb-targeted quantum dot (QD) bioconjugate and Kv1.3 blocking peptide improve metabolic health in obese male mice. J. Neurochem. 157, 1876–1896 (2021).

    CAS  PubMed  Google Scholar 

  26. Riera, C. E. et al. The sense of smell impacts metabolic health and obesity. Cell Metab. 26, 198–211 (2017).

    CAS  PubMed  Google Scholar 

  27. Cheng, D., Zhao, X., Yang, S., Cui, H. & Wang, G. Metabolomic signature between metabolically healthy overweight/obese and metabolically unhealthy overweight/obese: a systematic review. Diabetes Metab. Syndr. Obes. 14, 991–1010 (2021).

    PubMed  PubMed Central  Google Scholar 

  28. Blazing, R. M. & Franks, K. M. Odor coding in piriform cortex: mechanistic insights into distributed coding. Curr. Opin. Neurobiol. 64, 96–102 (2020).

    CAS  PubMed  Google Scholar 

  29. Horio, N., Murata, K., Yoshikawa, K., Yoshihara, Y. & Touhara, K. Contribution of individual olfactory receptors to odor-induced attractive or aversive behavior in mice. Nat. Commun. 10, 209 (2019).

    PubMed  PubMed Central  Google Scholar 

  30. Ferrero, D. M. et al. Detection and avoidance of a carnivore odor by prey. Proc. Natl Acad. Sci. USA 108, 11235–11240 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Denis, R. G. et al. Central orchestration of peripheral nutrient partitioning and substrate utilization: implications for the metabolic syndrome. Diabetes Metab. 40, 191–197 (2014).

    CAS  PubMed  Google Scholar 

  32. Castillo-Armengol, J., Fajas, L. & Lopez-Mejia, I. C. Inter-organ communication: a gatekeeper for metabolic health. EMBO Rep. 20, e47903 (2019).

    PubMed  PubMed Central  Google Scholar 

  33. Lee, J. et al. p38 MAPK-mediated regulation of Xbp1s is crucial for glucose homeostasis. Nat. Med. 17, 1251–1260 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Baiceanu, A., Mesdom, P., Lagouge, M. & Foufelle, F. Endoplasmic reticulum proteostasis in hepatic steatosis. Nat. Rev. Endocrinol. 12, 710–722 (2016).

    CAS  PubMed  Google Scholar 

  35. Kim, K. H. et al. Intermittent fasting promotes adipose thermogenesis and metabolic homeostasis via VEGF-mediated alternative activation of macrophage. Cell Res. 27, 1309–1326 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Møller, N. Ketone body, 3-hydroxybutyrate: minor metabolite — major medical manifestations. J. Clin. Endocrinol. Metab. 105, 2884–2892 (2020).

    Google Scholar 

  37. Palomer, X., Pizarro-Delgado, J., Barroso, E. & Vázquez-Carrera, M. Palmitic and oleic acid: the yin and yang of fatty acids in type 2 diabetes mellitus. Trends Endocrinol. Metab. 29, 178–190 (2018).

    CAS  PubMed  Google Scholar 

  38. Marangoni, F. et al. Dietary linoleic acid and human health: focus on cardiovascular and cardiometabolic effects. Atherosclerosis 292, 90–98 (2020).

    CAS  PubMed  Google Scholar 

  39. Fernandes-da-Silva, A. et al. Endoplasmic reticulum stress as the basis of obesity and metabolic diseases: focus on adipose tissue, liver, and pancreas. Eur. J. Nutr. 60, 2949–2960 (2021).

    CAS  PubMed  Google Scholar 

  40. Deem, J. D., Faber, C. L. & Morton, G. J. AgRP neurons: regulators of feeding, energy expenditure, and behavior. FEBS J. 289, 2362–2381 (2022).

    CAS  PubMed  Google Scholar 

  41. Chen, Y., Lin, Y. C., Zimmerman, C. A., Essner, R. A. & Knight, Z. A. Hunger neurons drive feeding through a sustained, positive reinforcement signal. eLife 5, e18640 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Huang, S., Xing, Y. & Liu, Y. Emerging roles for the ER stress sensor IRE1α in metabolic regulation and disease. J. Biol. Chem. 294, 18726–18741 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Tabas, I. & Ron, D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat. Cell Biol. 13, 184–190 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Bergman, B. C. & Goodpaster, B. H. Exercise and muscle lipid content, composition, and localization: influence on muscle insulin sensitivity. Diabetes 69, 848–858 (2020).

    CAS  PubMed  Google Scholar 

  45. Boone, M. H., Liang-Guallpa, J. & Krashes, M. J. Examining the role of olfaction in dietary choice. Cell Rep. 34, 108755 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Qiu, Q., Wu, Y., Ma, L. & Yu, C. R. Encoding innately recognized odors via a generalized population code. Curr. Biol. 31, 1813–1825 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Thiebaud, N. et al. Hyperlipidemic diet causes loss of olfactory sensory neurons, reduces olfactory discrimination, and disrupts odor-reversal learning. J. Neurosci. 34, 6970–6984 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    CAS  PubMed  Google Scholar 

  50. Adan, R. A. et al. The MC4 receptor and control of appetite. Br. J. Pharmacol. 149, 815–827 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Tsuneki, H. et al. Hypothalamic orexin prevents hepatic insulin resistance via daily bidirectional regulation of autonomic nervous system in mice. Diabetes 64, 459–470 (2015).

    CAS  PubMed  Google Scholar 

  52. Tsuneki, H. et al. Nighttime administration of nicotine improves hepatic glucose metabolism via the hypothalamic orexin system in mice. Endocrinology 157, 195–206 (2016).

    CAS  PubMed  Google Scholar 

  53. Soeda, Y. et al. The inositol phosphatase SHIP2 negatively regulates insulin/IGF-I actions implicated in neuroprotection and memory function in mouse brain. Mol. Endocrinol. 24, 1965–1977 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Tsuneki, H. et al. Different impacts of acylated and non-acylated long-acting insulin analogs on neural functions in vitro and in vivo. Diabetes Res. Clin. Pract. 129, 62–72 (2017).

    CAS  PubMed  Google Scholar 

  55. Fujita, K., Iguchi, Y., Une, M. & Watanabe, S. Ursodeoxycholic acid suppresses lipogenesis in mouse liver: possible role of the decrease in β-muricholic acid, a farnesoid X receptor antagonist. Lipids 52, 335–344 (2017).

    CAS  PubMed  Google Scholar 

  56. Sankoda, A. et al. Long-chain free fatty acid receptor GPR120 mediates oil-induced GIP secretion through CCK in male mice. Endocrinology 158, 1172–1180 (2017).

    CAS  PubMed  Google Scholar 

  57. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    PubMed  PubMed Central  Google Scholar 

  58. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  60. Ge, S. X., Son, E. W. & Yao, R. iDEP: an integrated web application for differential expression and pathway analysis of RNA-seq data. BMC Bioinf. 19, 534 (2018).

    CAS  Google Scholar 

  61. Gulshan, M. et al. Overexpression of Nmnat3 efficiently increases NAD and NGD levels and ameliorates age-associated insulin resistance. Aging Cell 17, e12798 (2018).

    PubMed  PubMed Central  Google Scholar 

  62. Okabe, K. et al. NAD+ metabolism regulates preadipocyte differentiation by enhancing α-ketoglutarate-mediated histone H3K9 demethylation at the PPARγ promoter. Front. Cell Dev. Biol. 8, 586179 (2020).

    PubMed  PubMed Central  Google Scholar 

  63. Wada, T. et al. Cilostazol ameliorates systemic insulin resistance in diabetic db/db mice by suppressing chronic inflammation in adipose tissue via modulation of both adipocyte and macrophage functions. Eur. J. Pharmacol. 707, 120–129 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was supported by JSPS KAKENHI (Grant Numbers JP18K08468, JP19H05011, JP19K08997, JP21K19704, and JP22H03506), Japan Diabetes Foundation (H. T. and T. S.), Smoking Research Foundation (H. T.), Toyama Pharmaceutical Valley Development Consortium (T. S.), Tamura Science and Technology Foundation (H. T.), First Bank of Toyama Scholarship Foundation Research Grant (H. T.), JST—the establishment of university fellowship towards the creation of science technology innovation (Grant Number JPMJFS2115, M. S.), and JST Moonshot R&D (Grant Number JPMJMS2021, T. S.).

Author information

Authors and Affiliations

Authors

Contributions

H. T. designed the experiments. M. S., T. I., K. S., H. M., K. O., K. Yubune, Y. M., S. N., T. Yamagishi, T. M., K. H., A. O., S. W., K. Yaku, D. O. and R. O. performed the experiments. H. T., M. N., K. I., T. N., T. W., T. Yasui and T. S. provided supervision. H. T. and T. S. wrote the manuscript with editing by all authors.

Corresponding authors

Correspondence to Hiroshi Tsuneki, Teruhito Yasui or Toshiyasu Sasaoka.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Increase in serum NEFA by food odor-stimulation in a fasting duration-dependent manner.

a,b, Influence of the fasting duration (a, 6 h vs. 24 h fasting; b, 16 h and 24 h fasting) on the olfactory exploratory behaviors of C57BL/6 J mice in the SOS4 cage with tubes releasing the already- known odors of normal chow diet (NCD), high fat diet (HFD) and high sucrose diet (HSD), and an empty tube (None). The time spent exploring each tube was evaluated. n = 10 biological replicates. Different letters (A, B, and C) represent significant difference based on two-way ANOVA followed by an ANOVA with Tukey’s test in a and by Bonferroni’s test in b. c, Five independent tests to analyze the changes in serum NEFA levels by NCD-odor stimulation in C57BL/6 J mice fasted for 24 h. The respective data are expressed as changes in absolute NEFA levels over time (upper), absolute difference (∆) in NEFA levels from baseline (middle), and percentage changes in NEFA levels from baseline (lower). n = 6–12 biological replicates (as indicated). Overall averages and the standard errors of these five results (same dataset) are shown in Fig. 2b. d,e, Serum NEFA levels during stimulation with or without NCD odor in the 6 h (d) and 16 h fasting state (e) in C57BL/6 J mice. n = 6 biological replicates. f, Apparatus used for forced olfactory stimulation (FOS1) equipped with an air-pump. The tube contained either NCD, HFD, HSD, or none. The odor release from the tube was assisted by an air-pump. g, Serum levels of NEFA (left panel) and β-hydroxybutyrate (right panel, β-OHB) in C57BL/6 J mice that were fasted for 24 h and stimulated with NCD odor in the FOS1 cage. n = 11 (None) and n = 12 (Odor) biological replicates. *P < 0.05, **P < 0.01 in c-g determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 2 The composition of serum NEFA during food-odor stimulation in the fasting state.

C57BL/6 J mice were fasted for 24 h, and stimulated with NCD odor for 0, 30, and 60 min in the SOS1 cage. The composition of serum NEFA was analyzed by gas-liquid chromatography. a, Time course of changes in the levels of palmitic acid, palmitoleic acid, stearic acid, vaccenic acid, oleic acid, linoleic acid, α-linoleic acid, arachidonic acid, and docosahexaenoic acid during the olfactory stimulation. Values represent differences from the basal concentration (that is, 0-min level) of each free fatty acid. b, Ratios of palmitic acid (PA) to oleic acid (OA), and those of stearic acid (SA) to OA. The ratio was normalized to the value at 0 min in each mouse, and averaged. n = 6 biological replicates. *P < 0.05, **P < 0.01 determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 3 DREADD-mediated derangement of the olfactory memory system impairs food odor-induced lipid mobilization during fasting.

a, Total cell numbers and infection success rates of pAAV-CaMKIIa-hM4Di-mCherry within layer I, II, and III in the piriform cortex (PC) of C57BL/6 J mice. Open columns indicate total numbers of Hoechst positive cells in each layer, whereas black columns indicate the numbers of mCherry positive cells. *P < 0.05 vs. total cell numbers in the layer II. #P < 0.05 vs. mCherry+ cell numbers in the layer II. Statistical significance was determined by one-way ANOVA with Dunnett’s test. n = 6 biological replicates. b, cFos expression after treatment with saline (SAL) or CNO followed by intermittent NCD-odor stimulations in the PC of fasted mice expressing hM4Di-mCherry. Left, representative images of cFos-like immunoreactivity (green), mCherry (violet) and Hoechst (blue) at 10X magnification. Scale bar = 200 μm. Inset, higher (63X) magnification images. White arrow head: cFos+mCherry+ cells. Black arrow head: cFos+mCherry cells. Scale bar = 50 μm. Right, probability for observing mCherry and cFos double positive cells (Prob. of cFos+mCherry+, closed column) compared to chance level (open column). *P < 0.05 vs. chance level, determined by two-sided Mann–Whitney U test. n = 4 biological replicates. c-f, Influence of DREADD-mediated derangement of the olfactory memory-processing system on food-searching behavior (c and e) and serum NEFA levels (d and f) during fasting in C57BL/6 J mice (12 months old) expressing hM4Di in the PC (c and d) and sham-operated controls (e and f). The effects of saline and CNO on the time spent exploring tubes which were releasing none, NCD, or already-known HFD odor in the SOS4 cage (c), and on the NEFA levels during exposure to HFD odor in the FOS1 cage (d) were compared. Different letters (A, B, and C) in c and e represent significant difference based on two-way ANOVA followed by an ANOVA with Tukey’s test in c and Bonferroni’s test in e. **P < 0.01 in d determined by paired two-tailed t-test. n = 12 biological replicates (c and d). n = 9 biological replicates (e and f). g, No correlation between the mCherry positive areas in the PC and the NEFA levels after 60 min of treatment with saline (upper) or CNO (lower) in mice expressing hM4Di-mCherry, determined by two-sided Pearson correlation coefficient test. n = 12 biological replicates. h-j, No influence of CNO on behaviors in the novel object recognition test (h, NOR), Y-maze test (i), and elevated plus maze test (j, EPM). n = 11 biological replicates. *P < 0.05 in h determined by one-way ANOVA with Dunnett’s test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 4 RNA-seq analysis of gene expression profiles in mice stimulated with food odor during fasting.

C57BL/6 J mice fasted for 24 h were stimulated with or without NCD odor for 1 h in the SOS1 cage. Gene expression profiles were determined by RNA-seq data analysis. a, Principal component analysis (PCA) of RNA-seq data for the hypothalamus, epididymal white adipose tissue (eWAT), small intestine (gut), soleus muscle, liver, and pancreas in odor-stimulated and control mice (n = 1 for each). b, Lists of DEGs in the transcriptome data of the soleus muscle and eWAT between the odor-stimulated and control mice. These DEGs were identified at a false discovery rate (FDR) < 0.05 and fold change > 1.5 (n = 3 biological replicates). The same DEGs between the soleus muscle and eWAT are highlighted in orange. c, Parametric analysis of gene set enrichment (PGGE) for group-on-group comparison into one-on-one comparison between DEGs and the related Gene Ontology (GO) biological process annotations of eWAT, pancreas (Panc), and soleus muscle in the odor-stimulated and control mice. The significantly different GO terms (P < 0.05) in terms of eWAT, pancreas, and soleus muscle transcriptome data between the odor-stimulated and control mice are labeled with †, §, and #, respectively.

Extended Data Fig. 5 Effects of non-food odors on serum NEFA levels.

a-d, Effects of attractive (muscone) and aversive odorants (PEA and TMT) on serum NEFA levels during fasting in C57BL/6 J mice. a, Timeline of the experimental protocol in b-d. Mice were fasted for 24 h, acclimated (Acc.) to the test cage in the absence of olfactory cue, and exposed to non-food odors in the FOS1 cage for 1 h. b, The effect of muscone. n = 6 biological replicates. c, The effect of PEA. n = 6 biological replicates. d, The effect of TMT. n = 6 biological replicates. e,f, Analysis of aversive behaviors in mice exposed to PEA and TMT during fasting in C57BL/6 J mice. e, The SOS2 apparatus used for the analysis of aversive behaviors. Two dishes with small holes were diagonally placed in the SOS2 cage. Non-food odorant was applied to a soft paper in one dish, and none was applied to another dish (a negative control). Mice fed ad libitum (AL) before the start of the tests were placed inside the SOS2 cage for 3 min, and time staining on half side with non-food-odorant was compared with that on the other half side with none. f, Upper: timeline of the experimental protocol. Lower: the effects of PEA (left, n = 8 biological replicates) and TMT (right, n = 6 biological replicates) on place preference between the non-food-odorant and control side. *P < 0.05 determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 6 Influences of food-odor stimulation during fasting on glucose and energy metabolism and gallbladder function in C57BL/6 J mice.

a, The gallbladder volumes 15 min (left) or 30 min (right) after oral fat administration. C57BL/6 J mice fasted for 24 h were prestimulated with NCD odor or none for 1 h, and then administered with corn oil or saline (SAL, 10 mL/kg). n = 7 (None-SAL) and n = 8 (None-OIL and Odor-OIL) biological replicates (in left). n = 10 biological replicates (in right). *P < 0.05 determined by one-way ANOVA with Dunnett’s test. b,c, Pyruvate tolerance test (b, PTT) and oral glucose tolerance test (c, OGTT) after food-odor stimulation during fasting. C57BL/6 J mice previously challenged with HFD were fasted for 24 h, and stimulated with or without HFD odor for 1 h in the SOS1 cage. Pyruvate (2 g/kg, i,p., n = 8 biological replicates) or glucose [3 g/kg, n = 7 (None) and n = 8 (Odor) biological replicates] was administered at 0 min. d,e, Serum levels of insulin (d) and adiponectin (e) in C57BL/6 J mice fasted for 24 h, stimulated with or without NCD odor in the SOS1 cage for 1 h, and then refed NCD. N.D.: not detected. n = 11 (None) and n = 13 (Odor) biological replicates. f,g, Energy expenditure (f) and locomotor activity (g) in saline-treated mice expressing hM4Di in the PC during fasting, stimulation with NCD odor, and subsequent refeeding of NCD. These data were obtained simultaneously with RQ in Fig. 6g. n = 8 biological replicates. h, i, Food intake in mice expressing hM4Di in the PC, that were fasted for 24 h, pretreated with saline (h, SAL, n = 8 biological replicates) or CNO (i, n = 7 (CNO-None) and n = 5 (CNO-Odor) biological replicates), stimulated with NCD odor for 1 h, and then refed NCD. The amounts of food intake in the refeeding state are shown. *P < 0.05 in b-i determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 7 Influences of food-odor stimulation during fasting on postprandial substrate utilization in orexin knockout and dopamine D2 receptor knockout mice.

a,b, Respiratory quotient (RQ) in orexin knockout (Orexin-/-) mice and their wild-type (Orexin+/+) mice (a) or dopamine D2 receptor knockout (D2R-/-) and their wild-type (D2R+/+) mice (b) that were fasted for 24 h, stimulated with the already-known food odor for 1 h, and then refed. n = 3 biological replicates. *P < 0.05 determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 8 Shift in balance of AgRP and Pomc gene expression in the hypothalamus of mice stimulated with food odor during fasting.

C57BL/6 J mice were fasted for 24 h, and stimulated with or without NCD odor for 1 h in the SOS1 cage. The expression levels of AgRP and Pomc mRNAs in the hypothalamus were measured by RT-PCR. a, Relative expression levels of AgRP and Pomc mRNAs compared to 18 S rRNA. b, The AgRP/Pomc expression ratio. c, The mRNA levels of hypothalamic neuropeptides, Orexin (Hypocretin), melanin-concentrating hormone (Mch), and thyrotropin-releasing hormone (Trh), compared to 18 S rRNA. n = 12 biological replicates. *P < 0.05, **P < 0.01 determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 9 Negligible effects of food-odor stimulation during fasting on metabolic activity in the adipose tissues and brain and gene expression in the liver after refeeding.

C57BL/6 J mice fasted for 24 h were stimulated with NCD odor or none for 1 h, and then refed NCD for 3 h. a, Metabolomic profiles in the epididymal white adipose tissue (eWAT), brown adipose tissue (BAT), and whole brain under the 3-h refeeding condition. OAA: oxaloacetic acid, α-KG: α-ketoglutarate. n = 8 (None) and n = 9 (Odor) biological replicates. b, Insulin tolerance test (ITT) under the 3-h refeeding condition. Insulin (0.5 unit/kg, i.p.) was injected at 0 min. n = 7 biological replicates. c, Expression levels of genes related to de novo lipogenesis and cholesterol synthesis in the liver under the 3-h refeeding condition. Srebp1c: sterol regulatory element-binding protein 1c, Acc: acetyl-CoA carboxylase, Fasn: fatty acid synthase, Mtp: microsomal triglyceride transfer protein, Atgl: adipose triglyceride lipase. n = 16 (None) and n = 18 (Odor) biological replicates. **P < 0.01 determined by unpaired two-tailed t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 10 Plausible mechanism underlying the link between food-odor perception and lipid metabolism.

Food-odor perception during fasting elicits biphasic regulations of lipid metabolism, as follows: Food-odor information is transmitted to the olfactory bulb-piriform cortex pathway, and processed for evaluating the valence, based on its memory. If the odor valence is found to be attractive during fasting, lipid mobilization was elicited as an acute-phase response through a rapid activation of hypothalamic POMC and sympathetic nervous system that promotes lipolysis in the white adipose tissue (WAT). Such lipid mobilization is considered to be beneficial for survival during starvation. Then, if the food is not immediately available, the late-phase responses are elicited through sustained activation of the hypothalamic AgRP signaling associated with reduction of sympathetic tones that can promote whole-body lipid utilization upon subsequent refeeding. Tissue-specific regulations of lipid metabolism, including an increase in the gastrointestinal (GI) lipid absorption, increase in the hepatic cholesterol turnover, decrease in the WAT lipolysis, and decrease in the skeletal muscle (SM) lipid metabolism seem to underlie the olfactory system-induced lipid partitioning. In particular, a liver-specific increase in metabolic flow and reduction of hepatic ER stress could contribute to the maintenance of metabolic health. NEFA: non-esterified fatty acid, CM: chylomicron, VLDL: very low-density lipoprotein.

Supplementary information

Supplementary Information

Supplementary Figures 1–9 and Supplementary Tables 1 and 2

Reporting Summary

Source data

Source Data Fig. 1

Numerical/Statistical Source Data

Source Data Fig. 2

Numerical/Statistical Source Data

Source Data Fig. 3

Numerical/Statistical Source Data

Source Data Fig. 3

Unprocessed microscopic images

Source Data Fig. 4

Numerical/Statistical Source Data

Source Data Fig. 5

Numerical/Statistical Source Data

Source Data Fig. 5

Unprocessed Western Blots and gels

Source Data Fig. 6

Numerical/Statistical Source Data

Source Data Fig. 7

Numerical/Statistical Source Data

Source Data Fig. 7

Unprocessed Western Blots and gels

Source Data Fig. 8

Numerical/Statistical Source Data

Source Data Extended Data Fig. 1

Numerical/Statistical Source Data

Source Data Extended Data Fig. 2

Numerical/Statistical Source Data

Source Data Extended Data Fig. 3

Numerical/Statistical Source Data

Source Data Extended Data Fig. 3

Unprocessed microscopic images

Source Data Extended Data Fig. 5

Numerical/Statistical Source Data

Source Data Extended Data Fig. 6

Numerical/Statistical Source Data

Source Data Extended Data Fig. 7

Numerical/Statistical Source Data

Source Data Extended Data Fig. 8

Numerical/Statistical Source Data

Source Data Extended Data Fig. 9

Numerical/Statistical 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

Tsuneki, H., Sugiyama, M., Ito, T. et al. Food odor perception promotes systemic lipid utilization. Nat Metab 4, 1514–1531 (2022). https://doi.org/10.1038/s42255-022-00673-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00673-y

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