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:

Sensory representation and detection mechanisms of gut osmolality change

Abstract

Ingested food and water stimulate sensory systems in the oropharyngeal and gastrointestinal areas before absorption1,2. These sensory signals modulate brain appetite circuits in a feed-forward manner3,4,5. Emerging evidence suggests that osmolality sensing in the gut rapidly inhibits thirst neurons upon water intake. Nevertheless, it remains unclear how peripheral sensory neurons detect visceral osmolality changes, and how they modulate thirst. Here we use optical and electrical recording combined with genetic approaches to visualize osmolality responses from sensory ganglion neurons. Gut hypotonic stimuli activate a dedicated vagal population distinct from mechanical-, hypertonic- or nutrient-sensitive neurons. We demonstrate that hypotonic responses are mediated by vagal afferents innervating the hepatic portal area (HPA), through which most water and nutrients are absorbed. Eliminating sensory inputs from this area selectively abolished hypotonic but not mechanical responses in vagal neurons. Recording from forebrain thirst neurons and behavioural analyses show that HPA-derived osmolality signals are required for feed-forward thirst satiation and drinking termination. Notably, HPA-innervating vagal afferents do not sense osmolality itself. Instead, these responses are mediated partly by vasoactive intestinal peptide secreted after water ingestion. Together, our results reveal visceral hypoosmolality as an important vagal sensory modality, and that intestinal osmolality change is translated into hormonal signals to regulate thirst circuit activity through the HPA pathway.

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: The vagus nerve responds to visceral osmolality changes.
Fig. 2: Visceral hypotonic stimuli activate a dedicated vagal population.
Fig. 3: Vagal sensory inputs from the HPA transmits gut-to-brain osmolality signals.
Fig. 4: HPA vagal afferents are required for pre-absorptive thirst satiation.
Fig. 5: Visceral osmolality sensing recruits VIP–VIPR2 signalling in the HPA.

Similar content being viewed by others

Data availability

Additional data that support the finding of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

The MATLAB code used to perform the imaging analysis is modified from the CalmAn code at https://github.com/flatironinstitute/CaImAn-MATLAB, and is available at https://github.com/Oka-Lab/Imaging-analysis.

References

  1. Browning, K. N., Verheijden, S. & Boeckxstaens, G. E. The vagus nerve in appetite regulation, mood, and intestinal inflammation. Gastroenterology 152, 730–744 (2017).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Andermann, M. L. & Lowell, B. B. Toward a wiring diagram understanding of appetite control. Neuron 95, 757–778 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Augustine, V., Lee, S. & Oka, Y. Neural control and modulation of thirst, sodium appetite, and hunger. Cell 180, 25–32 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zimmerman, C. A., Leib, D. E. & Knight, Z. A. Neural circuits underlying thirst and fluid homeostasis. Nat. Rev. Neurosci. 18, 459–469 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Augustine, V. et al. Hierarchical neural architecture underlying thirst regulation. Nature 555, 204–209 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ichiki, T., Augustine, V. & Oka, Y. Neural populations for maintaining body fluid balance. Curr. Opin. Neurobiol. 57, 134–140 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zimmerman, C. A. & Knight, Z. A. Layers of signals that regulate appetite. Curr. Opin. Neurobiol. 64, 79–88 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lowell, B. B. New neuroscience of homeostasis and drives for food, water, and salt. N. Engl. J. Med. 380, 459–471 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Augustine, V., Gokce, S. K. & Oka, Y. Peripheral and central nutrient sensing underlying appetite regulation. Trends Neurosci. 41, 526–539 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Thrasher, T. N., Nistal-Herrera, J. F., Keil, L. C. & Ramsay, D. J. Satiety and inhibition of vasopressin secretion after drinking in dehydrated dogs. Am. J. Physiol. 240, E394–E401 (1981).

    CAS  PubMed  Google Scholar 

  12. Zimmerman, C. A. et al. A gut-to-brain signal of fluid osmolarity controls thirst satiation. Nature 568, 98–102 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Augustine, V. et al. Temporally and spatially distinct thirst satiation signals. Neuron 103, 242–249.e244 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lechner, S. G. et al. The molecular and cellular identity of peripheral osmoreceptors. Neuron 69, 332–344 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Brierley, S. M. et al. Selective role for TRPV4 ion channels in visceral sensory pathways. Gastroenterology 134, 2059–2069 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Zimmer, L. J., Meliza, L. & Hsiao, S. Effects of cervical and subdiaphragmatic vagotomy on osmotic and volemic thirst. Physiol. Behav. 16, 665–670 (1976).

    Article  CAS  PubMed  Google Scholar 

  17. Smith, G. P. & Jerome, C. Effects of total and selective abdominal vagotomies on water intake in rats. J. Auton. Nerv. Syst. 9, 259–271 (1983).

    Article  CAS  PubMed  Google Scholar 

  18. Kim, D. Y. et al. A neural circuit mechanism for mechanosensory feedback control of ingestion. Nature 580, 376–380 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. 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 

  20. Kaelberer, M. M. et al. A gut–brain neural circuit for nutrient sensory transduction. Science 361, eaat523 (2018).

    Article  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kupari, J., Haring, M., Agirre, E., Castelo-Branco, G. & Ernfors, P. An atlas of vagal sensory neurons and their molecular specialization. Cell Rep. 27, 2508–2523.e2504 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bai, L. et al. Genetic identification of vagal sensory neurons that control feeding. Cell 179, 1129–1143.e1123 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Prescott, S. L., Umans, B. D., Williams, E. K., Brust, R. D. & Liberles, S. D. An airway protection program revealed by sweeping genetic control of vagal afferents. Cell 181, 574–589.e514 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Adachi, A., Niijima, A. & Jacobs, H. L. An hepatic osmoreceptor mechanism in the rat: electrophysiological and behavioral studies. Am. J. Physiol. 231, 1043–1049 (1976).

    Article  CAS  PubMed  Google Scholar 

  26. Baertschi, A. J. & Vallet, P. G. Osmosensitivity of the hepatic portal vein area and vasopressin release in rats. J. Physiol. 315, 217–230 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Morita, H., Fujiki, N., Hagiike, M., Yamaguchi, O. & Lee, K. Functional evidence for involvement of bumetanide-sensitive Na+K+2Cl cotransport in the hepatoportal Na+ receptor of the Sprague–Dawley rat. Neurosci. Lett. 264, 65–68 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Niijima, A. Glucose-sensitive afferent nerve fibres in the hepatic branch of the vagus nerve in the guinea-pig. J. Physiol. 332, 315–323 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. McKinley, M. J. & Johnson, A. K. The physiological regulation of thirst and fluid intake. News Physiol Sci 19, 1–6 (2004).

    PubMed  Google Scholar 

  30. Gribble, F. M. & Reimann, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat. Rev. Endocrinol. 15, 226–237 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Christofides, N. D. et al. Release of gastrointestinal hormones following an oral water load. Experientia 35, 1521–1523 (1979).

    Article  CAS  PubMed  Google Scholar 

  32. Costa, M. & Furness, J. B. The origins, pathways and terminations of neurons with VIP-like immunoreactivity in the guinea-pig small intestine. Neuroscience 8, 665–676 (1983).

    Article  CAS  PubMed  Google Scholar 

  33. Talbot, J. et al. Feeding-dependent VIP neuron–ILC3 circuit regulates the intestinal barrier. Nature 579, 575–580 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Furness, J. B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 9, 286–294 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. 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 

  36. Kennedy, A. et al. Stimulus-specific hypothalamic encoding of a persistent defensive state. Nature 586, 730–734 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zocchi, D., Wennemuth, G. & Oka, Y. The cellular mechanism for water detection in the mammalian taste system. Nat. Neurosci. 20, 927–933 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Xie, C., Wei, W., Zhang, T., Dirsch, O. & Dahmen, U. Monitoring of systemic and hepatic hemodynamic parameters in mice. J. Vis. Exp. 92, e51955 (2014).

    Google Scholar 

  39. McHugh, J. et al. Portal osmopressor mechanism linked to transient receptor potential vanilloid 4 and blood pressure control. Hypertension 55, 1438–1443 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Lerner, T. N. et al. Intact-brain analyses reveal distinct information carried by SNc dopamine subcircuits. Cell 162, 635–647 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Han, W. et al. A neural circuit for gut-induced reward. Cell 175, 665–678.e623 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hama, H. et al. ScaleS: an optical clearing palette for biological imaging. Nat. Neurosci. 18, 1518–1529 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. L’Heureux-Bouron, D. et al. Total subdiaphragmatic vagotomy does not suppress high protein diet-induced food intake depression in rats. J. Nutr. 133, 2639–2642 (2003).

    Article  PubMed  Google Scholar 

  44. Lee, S. et al. Chemosensory modulation of neural circuits for sodium appetite. Nature 568, 93–97 (2019).

Download references

Acknowledgements

We thank the members of the Oka laboratory, S. D. Liberles and C. S. Zuker for helpful discussion and comments; B. Ho and A. Koranne for maintaining and genotyping mouse lines; T. Zhang, W. Han and I. E. Araujo for technical advice on surgical techniques; L. Luebbert for initial imaging analysis; T. Karigo for technical help; X. Chen and V. Gradinaru for advice on virus development and tissue clearing; and J. Parker for sharing the qPCR machine. This work was supported by Startup funds from the President and Provost of California Institute of Technology and the Biology and Biological Engineering Division of California Institute of Technology. Y.O. is also supported by New York Stem Cell Foundation, NIH (R01NS109997, R01NS123918), Alfred P. Sloan Foundation, and Heritage Medical Research Institute. T.I. is supported by the Japan Society for the Promotion of Science.

Author information

Authors and Affiliations

Authors

Contributions

T.I. and Y.O. conceived the research programme and designed experiments. T.I. performed the experiments and analysed the data, with help from T.W. A.K., and D.J.A. wrote the code for the imaging analysis and provided advice on the data analysis. A.-H.P. analysed the single-cell RNA sequencing data. H.E. performed and analysed chemogenetic experiments. T.I. and Y.O. wrote the paper. Y.O. supervised the entire work.

Corresponding author

Correspondence to Yuki Oka.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review information

Nature thanks Richard Palmiter and the other, anonymous reviewers 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 Responses of vagal and thoracic DRG neurons in vivo.

a, Schematic of intestinal infusion during electrophysiological and optical recording. Isotonic (0.9%) saline was continuously perfused using a peristaltic pump. Stimulus was delivered using a solenoid valve at the same flow rate (800 μL/min). b, Integrated vagal nerve responses during stimulation (AUC) were quantified (n = 8 mice for saline and water, and n = 6 for other stimuli). c, Quantification of vagal water response at different flow rates (n = 5 mice). d, Number of WGA-positive cells in DRG (thoracic,T5-lumber,L3) retrogradely tracing from HPA, intestine or stomach. Representative images from 3 mice for each tracing site are shown in the right. Scale bar, 100 μm. e, Schematic diagram of DRG imaging. GRIN lens was placed on a thoracic DRG (T12 or 13). Calcium dynamics of individual neurons was visualized in Slc17a6-Cre;Ai96 mice. f, Response heatmaps to skin pinch, intestinal infusion of 4% acetic acid, water, 1M NaCl and Ensure from individual DRG neurons (ΔF/F(σ), left) and percentage of responding neurons (right, n = 3 mice for acetic acid, water and NaCl, 4 mice for other stimuli). *P < 0.05, **P < 0.01, ***P < 0.001 by Kruskal–Wallis test (with Dunn’s post test). Data are shown as mean ± s.e.m.

Source data

Extended Data Fig. 2 Osmolality and mechanical responses under vagal ganglia imaging.

a, Calcium dynamics during saline (blue), first (red) and second (purple) intestinal water infusion (Slc17a6-Cre;Ai96 mice). Representative images and a heatmap of individual vagal responses from 4 animal are shown. 64% of neurons activated by one trial were also activated by another trial. b, After subdiaphragmatic vagotomy (SVx), responses to intestinal water, 500 mM NaCl infusion, and distension were abolished (left). The number of total responding neurons was quantified (right, Slc17a6-Cre;Ai96, n = 4 mice). c, Heatmap of individual vagal activity during short- (20 s) and long- (2 min) term of intestinal water infusion (Slc17a6-Cre;Ai96, n = 3 mice). d, Osmotic-stimuli and mechanical distension activated largely non-overlapping neurons. Venn diagrams indicate the number of responding neurons (right, Slc17a6-Cre;Ai96, n = 3 mice). e, Representative vagal ganglia responses to water infusion and distension in the intestine (red) and stomach (green, n = 4 mice). f, Total number of vagal neurons responding to the indicated stimuli in the intestine (red, n = 4 mice) and stomach (green, n = 4 mice). Scale bar, 200 μm. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 3 Transcriptomic analyses of the vagal ganglia.

a, Vagal ganglia contain 27 transcriptomic neuron types shown in a t-SNE embedding with color-coded cell identity. b, Violin plot of log-normalized expression of marker genes in vagal ganglia, including Tac1 and Nts with maximum counts per million (max CPM) above. t-SNE embedding for Nts (c) and Tac1 (d) shows log-normalized expression in vagal neuron types. These data are reanalyzed and plotted from a previous study (Prescott et al, 2020)24. e, Two-color RNA in situ hybridization for validating Cre expression in Tac1-Cre lines. 91.2% of Tac1-Cre expression matched endogenous gene expression. Scale bar, 20 µm.

Extended Data Fig. 4 Functional manipulation of TAC1+ and NTS+ vagal neurons.

a, TAC1+ neurons that responded to intestinal water or 500 mM NaCl are shown (Tac1-Cre;Ai162D, n = 4 mice). Same datasets from Fig. 3a are shown. b, Chemogenetic activation of TAC1+ vagal neurons significantly inhibited water intake after deprivation. AAV-hSyn-DIO-hM3D(Gq)-mCherry was injected into the vagal ganglia of Nts-Cre, Tac1-Cre, Slc17a6-Cre, or wild type (WT) mice. Water intake after 24-h water deprivation, 300 mM sucrose or chow consumption after 24-h food deprivation were measured after intraperitoneal PBS (vehicle) or clozapine N-oxide (CNO) administration (n = 7, 7, 5, and 4 mice (water and sucrose), and 5, 5, 4, and 4 mice (chow) were analyzed for Nts-Cre, Tac1-Cre, Slc17a6-Cre and WT animals, respectively.). c, Loss-of-function of TAC1+ vagal neurons. Cre-dependent taCasp3-encoding AAV was injected into the vagal ganglia of Tac1-Cre mice (left, n = 6) or WT mice (right, n = 6). After 2 weeks of recovery period, water intake was measured under water-deprived state. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant by two-way ANOVA (Šídák multiple comparisons) or two-tailed paired t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 5 Anatomical projection patterns of TAC1+ and NTS+ vagal sensory neurons.

Whole-mount cleared tissue or sliced brain sections showing vagal terminals (AAV-Flex-tdTomato, red) and cell bodies (counterstained with DAPI or NeuN for Vagal ganglia and DRG, blue). Enteric neurons are labeled by Fluoro-Gold (green). A magnified image of stomach IGLE in a Slc17a6-Cre mouse is presented (bottom, left). The expression patterns in Slc17a6-Cre, Tac1-Cre and Nts-Cre mice are summarized (bottom, right, n = 3 animals for each mouse line). NTS, nucleus of the solitary tract; CG-SMG, celiac ganglion and superior mesenteric ganglion complex. Scale bar, 200 μm.

Extended Data Fig. 6 Anatomical projection patterns of vagal sensory neurons in the liver.

a, Whole-mount staining of innervation of SLC17A6+, TAC1+, NTS+ vagal populations in the hepatic area (n = 2 animals for each mouse line). Sensory terminals were visualized by AAV-Flex-tdTomato (red). Scale bar, 100 μm. b, Cell bodies of vagal sensory neurons were retrogradely labeled by WGA 555 (magenta) from the different liver parts (#2, #4, or all parts except #2 and #4, referred from a), and by WGA 647 (green) from HPV. The vagal innervation to the hilus and HPV mostly overlapped due to their proximity (84%), while the majority of the liver lacked innervation except a limited area in the left lobe (n = 3 mice). Scale bar, 200 μm. c, Hepatic branch denervation with intact liver branch abolished intestinal hypoosmotic responses (Slc17a6-Cre;Ai96, n = 3 mice).

Extended Data Fig. 7 Hepatic-branch-specific vagal denervation and its physiological significance.

a, Heatmap of individual vagal activity during long-term intestinal water infusion before and after HVx (Slc17a6-Cre;Ai96, n = 3 mice). b, Distinct vagal populations project to different organs. Whole-mount vagal sensory neurons retrogradely labeled by WGA 555 (magenta) and WGA 647 (green) from the same organ (HPA vs HPA) or different organs (HPA vs intestine, HPA vs stomach). Percentage of overlapping neurons is quantified (right, n = 3 mice). c, Validation of hepatic-branch-specific denervation. Vagal sensory neurons were retrogradely labeled by WGA 555 from intestine (magenta) and WGA 647 from HPA (green). After HPA denervation, the number of labeled neurons from HPA was drastically reduced (n = 3 mice). d, Reversible inhibition of water responses by a local anesthesia to HPA. Vagal responses to intestinal water infusion before and after application of 5% lidocaine (left, Slc17a6-Cre;Ai96, n = 4 mice). Responses recovered after the drug wore off. The number of the responded cells and mean ΔF/F(σ) values in neurons responded after 100 min are shown in the bottom. By contrast, the same anesthesia treatment did not affect intestinal distension responses (right, Slc17a6-Cre;Ai96, n = 3 mice). e, Duodenum and pancreas denervation using 85% phenol did not affect vagal activity during intestinal water infusion (left, Slc17a6-Cre;Ai96, n = 2 mice). Denervation was verified by injecting WGA 555 to pancreas (magenta) and WGA 647 to duodenum (green). After denervation, the number of retrogradely labeled neurons was drastically reduced (right, n = 3 mice). Scale bar, 200 μm. *P < 0.05, ***P < 0.001, ns, not significant by two-way or one-way ANOVA (Šídák multiple comparisons) or by Friedman test (Dunn’s multiple comparisons). Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 8 Functional and anatomical analyses of the hepatic vagal branch.

a, Vagal responses to intestinal water stimuli were abolished after HPA denervation using 85% phenol (Slc17a6-Cre;Ai96, n = 4 mice). b and c, Vagal response heatmaps to 500 mM NaCl or 300 mM glucose stimuli before and after HVx (b, n = 3 mice), or after injecting WGA 647 into the HPA (c, n = 3 mice). d, A minor population of intestine-innervating neurons (labeled by WGA 647) responded to intestinal acute water stimulus (Slc17a6-Cre;Ai96, n = 3 mice).

Extended Data Fig. 9 The effects of vagal manipulation on nutrient consumption and SFO neuron activity.

a, Water intake after 24-h deprivation during a 20-min session after HVx (n = 9 mice for sham and HVx, left) and HPA denervation using 85% phenol (n = 4 for sham, n = 7 mice for denervation, right). The same dataset from Fig. 4a for HVx experiments is presented. b, No change was observed for daily ad libitum water intake in sham (grey) and HPA denervation (red) animals (n = 5 mice). c, The vagus nerve does not contribute to water absorption. Plasma osmolality was measured in water-deprived mice after intestinal water infusion with or without SVx (n = 3 for sham, n = 3 for water-deprived SVx mice, n = 4 for water-infused SVx mice). d, Water (left) or food (normal chow, middle) intake after 24-h deprivation during a 20-min (water) or 1-h (food) session. Consistent with previous studies17,43, no difference was observed between sham (grey) and SVx (red) mice, or pre- and post-operation (n = 5 and 11 mice for pre- and post-op water intake measurement, n = 5 mice for food consumption measurement). Spontaneous daily water intake under sated states was not affected (right, n = 5 mice). e, Representative stomach and intestine samples after SVx (left) or HVx (right), with sham as a comparison. Stomach weight was significantly increased in the SVx animals but not in the HVx animals (n = 5 mice for sham, n = 6 mice for SVx and HVx). f, A diagram of vagal sensory circuits transmitting thirst satiation signals. g, Simultaneous photometry recording from SFONOS1 neurons and chemogenetic stimulation of vagal ganglion neurons. AAV-hSyn-hM3D(Gq)-mCherry was injected into the vagal ganglia while SFONOS1 neurons were infected with AAV-Flex-GCaMP6s in Nos1-Cre animals. h, Calcium dynamics of thirst neurons after PBS (vehicle) or CNO administration (n = 5 mice). Chemogenetic activation of vagal ganglia induced strong feed-forward inhibition in thirst neurons in sated- (left, red trace) and water-deprived condition (right, red trace). AUC was quantified between 300 to 600 s after CNO injection. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant by two-way ANOVA (Šídák multiple comparisons), two-tailed Mann-Whitney U-test, or two-tailed paired t-test. Data are presented as mean ± s.e.m.

Source data

Extended Data Fig. 10 Contribution of hormones to HPA-innervating vagal responses and fluid balance.

a, Representative responses of vagal neurons to hormones infused into the HPV before (red) and after (green) HVx (Slc17a6-Cre;Ai96 mice, n = 3 mice). Scale bar, 200 μm. b, Vagal responses to intestinal water infusion and HPV GLP1 infusion (n = 3 mice). c, Vagal responses to a near-physiological concentration of VIP and intestinal hypoosmotic stimuli (n = 3 mice). d, VIP concentration in the HPV plasma was measured by ELISA from water-deprived (W.D.) animals and water-repleted animals with 10 min or 20 min water access (left, n = 9 mice for W.D., n = 8 mice for +water 10 min, and n = 6 mice for +water 20 min). No change in the systemic VIP level was observed in the retro-orbital sinus after water consumption (right, n = 9 mice for W.D., n = 8 mice for +water 10 min). e, After VIPR antagonist application, water and VIP dual-responding neurons were suppressed. f, qPCR analysis of VIPR1 and VIPR2 expression in the HPA, liver, duodenum, and colon. The same dataset from Fig. 5e is presented. *P < 0.05 by one-way ANOVA (Šídák multiple comparisons) or two-tailed unpaired t-test. Data are presented as mean ± s.e.m.

Source data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ichiki, T., Wang, T., Kennedy, A. et al. Sensory representation and detection mechanisms of gut osmolality change. Nature 602, 468–474 (2022). https://doi.org/10.1038/s41586-021-04359-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04359-5

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