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:

A neural basis for tonic suppression of sodium appetite

Abstract

Sodium appetite is a powerful form of motivation that can drive ingestion of high, yet aversive concentrations of sodium in animals that are depleted of sodium. However, in normal conditions, sodium appetite is suppressed to prevent homeostatic deviations. Although molecular and neural mechanisms underlying the stimulation of sodium appetite have received much attention recently, mechanisms that inhibit sodium appetite remain largely obscure. Here we report that serotonin 2c receptor (Htr2c)-expressing neurons in the lateral parabrachial nucleus (LPBNHtr2c neurons) inhibit sodium appetite. Activity of these neurons is regulated by bodily sodium content, and their activation can rapidly suppress sodium intake. Conversely, inhibition of these neurons specifically drives sodium appetite, even during euvolemic conditions. Notably, the physiological role of Htr2c expressed by LPBN neurons is to disinhibit sodium appetite. Our results suggest that LPBNHtr2c neurons act as a brake against sodium appetite and that their alleviation is required for the full manifestation of sodium appetite.

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: LPBNHtr2c neurons are regulated by bodily sodium content.
Fig. 2: Activation of LPBNHtr2c neurons suppresses sodium intake.
Fig. 3: Inhibition of LPBNHtr2c neurons increases sodium intake.
Fig. 4: Stimulation of the LPBNHtr2c → CeA projection suppresses sodium intake.
Fig. 5: Htr2c inhibits LPBN neurons via KATP channels.
Fig. 6: Htr2c in the LPBN is necessary to disinhibit sodium appetite during hypovolemia.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

Code availability

Custom code used in this study is available from the corresponding authors upon reasonable request.

References

  1. Wagman, W. Sodium chloride deprivation: development of sodium chloride as a reinforcement. Science 140, 1403–1404 (1963).

    Article  CAS  Google Scholar 

  2. Matsuda, T.et al. Distinct neural mechanisms for the control of thirst and salt appetite in the subfornical organ. Nat. Neurosci. 20, 230–241 (2017).

    PubMed  Google Scholar 

  3. Jarvie, B. C. & Palmiter, R. D. HSD2 neurons in the hindbrain drive sodium appetite. Nat. Neurosci. 20, 167–169 (2017).

    Article  Google Scholar 

  4. Resch, J. M. et al. Aldosterone-sensing neurons in the NTS exhibit state-dependent pacemaker activity and drive sodium appetite via synergy with angiotensin II signaling. Neuron 96, 190–206 (2017).

    Article  CAS  Google Scholar 

  5. Oka, Y., Butnaru, M., von Buchholtz, L., Ryba, N. J. & Zuker, C. S. High salt recruits aversive taste pathways. Nature 494, 472–475 (2013).

    Article  Google Scholar 

  6. de Oliveira, L. B., Callera, J. C., De Luca, L. A. Jr, Colombari, D. S. & Menani, J. V. GABAergic mechanisms of the lateral parabrachial nucleus on sodium appetite. Brain Res. Bull. 73, 238–247 (2007).

    Article  Google Scholar 

  7. De Gobbi, J. I. et al. Non-NMDA receptors in the lateral parabrachial nucleus modulate sodium appetite. Brain Res. 1301, 44–51 (2009).

    Article  Google Scholar 

  8. Johnson, A. K. & Thunhorst, R. L. The neuroendocrinology of thirst and salt appetite: visceral sensory signals and mechanisms of central integration. Front. Neuroendocrinol. 18, 292–353 (1997).

    Article  CAS  Google Scholar 

  9. Scalera, G., Spector, A. C. & Norgren, R. Excitotoxic lesions of the parabrachial nuclei prevent conditioned taste aversions and sodium appetite in rats. Behav. Neurosci. 109, 997–1008 (1995).

    Article  Google Scholar 

  10. Geerling, J. C. et al. FoxP2 expression defines dorsolateral pontine neurons activated by sodium deprivation. Brain Res. 1375, 19–27 (2011).

    Article  CAS  Google Scholar 

  11. Castro, L. et al. Central 5-HT2B/2C and 5-HT3 receptor stimulation decreases salt intake in sodium-depleted rats. Brain Res. 981, 151–159 (2003).

    Article  CAS  Google Scholar 

  12. Gentili, L., Saija, A., Luchetti, G. & Massi, M. Effect of the 5-HT2 antagonist ketanserin on salt appetite in the rat. Pharmacol. Biochem. Behav. 39, 171–176 (1991).

    Article  CAS  Google Scholar 

  13. Menani, J. V., Thunhorst, R. L. & Johnson, A. K. Lateral parabrachial nucleus and serotonergic mechanisms in the control of salt appetite in rats. Am. J. Physiol. 270, R162–R168 (1996).

    CAS  PubMed  Google Scholar 

  14. De Gobbi, J. I. et al. 5-HT2 and 5-HT3 receptors in the lateral parabrachial nucleus mediate opposite effects on sodium intake. Neuroscience 146, 1453–1461 (2007).

  15. Yamamoto, T. et al. c-Fos expression in the parabrachial nucleus after ingestion of sodium chloride in the rat. Neuroreport 4, 1223–1226 (1993).

    Article  CAS  Google Scholar 

  16. Geerling, J. C. & Loewy, A. D. Sodium deprivation and salt intake activate separate neuronal subpopulations in the nucleus of the solitary tract and the parabrachial complex. J. Comp. Neurol. 504, 379–403 (2007).

    Article  CAS  Google Scholar 

  17. Ryan, P. J., Ross, S. I., Campos, C. A., Derkach, V. A. & Palmiter, R. D. Oxytocin-receptor-expressing neurons in the parabrachial nucleus regulate fluid intake. Nat. Neurosci. 20, 1722–1733 (2017).

    Article  Google Scholar 

  18. Donnelly, M. L. et al. Analysis of the aphthovirus 2A/2B polyprotein ‘cleavage’ mechanism indicates not a proteolytic reaction, but a novel translational effect: a putative ribosomal ‘skip’. J. Gen. Virol. 82, 1013–1025 (2001).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  20. Campos, C. A. et al. Cancer-induced anorexia and malaise are mediated by CGRP neurons in the parabrachial nucleus. Nat. Neurosci. 20, 934–942 (2017).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Garfield, A. S. et al. A neural basis for melanocortin-4 receptor-regulated appetite. Nat. Neurosci. 18, 863–871 (2015).

    Article  Google Scholar 

  23. Stricker, E. M. & Verbalis, J. G. Central inhibitory control of sodium appetite in rats: correlation with pituitary oxytocin secretion. Behav. Neurosci. 101, 560 (1987).

    Article  CAS  Google Scholar 

  24. Geerling, J. C., Chimenti, P. C. & Loewy, A. D. Phox2b expression in the aldosterone-sensitive HSD2 neurons of the NTS. Brain Res. 1226, 82–88 (2008).

    Article  CAS  Google Scholar 

  25. Kang, B. J. et al. Central nervous system distribution of the transcription factor Phox2b in the adult rat. J. Comp. Neurol. 503, 627–641 (2007).

    Article  CAS  Google Scholar 

  26. Hashimoto, K., Obata, K. & Ogawa, H. Characterization of parabrachial subnuclei in mice with regard to salt tastants: possible independence of taste relay from visceral processing. Chem. Senses 34, 253–267 (2009).

    Article  Google Scholar 

  27. Biondolillo, J. W., Williams, L. A. & King, M. S. Blocking glutamate receptors in the waist area of the parabrachial nucleus decreases taste reactivity behaviors in conscious rats. Chem. Senses 34, 221–230 (2009).

    Article  CAS  Google Scholar 

  28. Chandrashekar, J. et al. The cells and peripheral representation of sodium taste in mice. Nature 464, 297–301 (2010).

    Article  Google Scholar 

  29. Zardetto-Smith, A. M., Beltz, T. G. & Johnson, A. K. Role of the central nucleus of the amygdala and bed nucleus of the stria terminalis in experimentally-induced salt appetite. Brain Res. 645, 123–134 (1994).

    Article  CAS  Google Scholar 

  30. Galaverna, O., De Luca, L. A. Jr, Schulkin, J., Yao, S. Z. & Epstein, A. N. Deficits in NaCl ingestion after damage to the central nucleus of the amygdala in the rat. Brain Res. Bull. 28, 89–98 (1992).

    Article  CAS  Google Scholar 

  31. Dong, H. W., Petrovich, G. D. & Swanson, L. W. Topography of projections from amygdala to bed nuclei of the stria terminalis. Brain Res. Rev. 38, 192–246 (2001).

    Article  CAS  Google Scholar 

  32. Dong, H. W., Petrovich, G. D., Watts, A. G. & Swanson, L. W. Basic organization of projections from the oval and fusiform nuclei of the bed nuclei of the stria terminalis in adult rat brain. J. Comp. Neurol. 436, 430–455 (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  34. Chen, Y., Baez, M. & Yu, L. Functional coupling of the 5-HT2C serotonin receptor to G proteins in Xenopus oocytes. Neurosci. Lett. 179, 100–102 (1994).

    Article  CAS  Google Scholar 

  35. Lucaites, V. L., Nelson, D. L., Wainscott, D. B. & Baez, M. Receptor subtype and density determine the coupling repertoire of the 5-HT2 receptor subfamily. Life Sci. 59, 1081–1095 (1996).

    Article  CAS  Google Scholar 

  36. Kurachi, Y., Tung, R. T., Ito, H. & Nakajima, T. G protein activation of cardiac muscarinic K+ channels. Prog. Neurobiol. 39, 229–246 (1992).

    Article  CAS  Google Scholar 

  37. Petrov, T., Krukoff, T. L. & Jhamandas, J. H. The hypothalamic paraventricular and lateral parabrachial nuclei receive collaterals from raphe nucleus neurons: a combined double retrograde and immunocytochemical study. J. Comp. Neurol. 318, 18–26 (1992).

    Article  CAS  Google Scholar 

  38. Bang, S. J., Jensen, P., Dymecki, S. M. & Commons, K. G. Projections and interconnections of genetically defined serotonin neurons in mice. Eur. J. Neurosci. 35, 85–96 (2012).

    Article  Google Scholar 

  39. Smits, J. F., van Essen, H. & Struyker-Boudier, H. A. Serotonin-mediated cardiovascular responses to electrical stimulation of the raphe nuclei in the rat. Life Sci. 23, 173–178 (1978).

    Article  CAS  Google Scholar 

  40. Berglund, E. D. et al. Serotonin 2C receptors in pro-opiomelanocortin neurons regulate energy and glucose homeostasis. J. Clin. Invest. 123, 5061–5070 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G.S.B. Suh (KAIST) for helpful comments on the manuscript, S.-H. Lee (KAIST) for technical help with viral tracing experiments and D. Kim (KAIST) for technical help with imaging. This work was supported by grants from the Samsung Science & Technology Foundation (SSTF-BA1901-11 to J.-W.S.), the KAIST Future Systems Health Care Project (to J.-W.S.), the KAIST Venture Research Program for Graduate and PhD students (to S.P.), the American Heart Association (16SDG27260001 to C.L.) and the National Institutes of Health (R01 DK114036 to C.L. and R01 DK100699, R01 DK119169, DK119130 5830 to K.W.W.). S.P. was supported in part by stipends from the National Research Foundation of Korea (NRF-2015M3A9E7029177 and NRF-2016R1C1B2006614 to J.-W.S.).

Author information

Authors and Affiliations

Authors

Contributions

S.P. and J.-W.S. designed the experiments. S.P. conducted experiments and analyzed data. C.L. and K.W.W. provided reagents and expertise. C.L. generated and validated Htr2c-2A-iCre mice. S.P. and J.-W.S. wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Chen Liu or Jong-Woo Sohn.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Yuki Oka 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 LPBN neurons respond to hypertonic saline.

Hypertonic saline-loaded mice show more Fos in the LPBN than saline-loaded controls. Two-tailed Mann-Whitney test, Saline N = 5 mice, Hypertonic saline N =5 mice. P = 0.0317. All data represented as mean ± s.e.m. *P < 0.05.

Extended Data Fig. 2 Generation of an Htr2c-2A-iCre mouse line.

(a) Htr2c-2A-iCre knock-in mice in which the coding sequences of Htr2c and iCre are linked by a viral peptide bridge 2A sequence. (b) Detection of wildtype and targeted Htr2c alleles by PCR genotyping. Similar results were obtained with at least 3 independent replications. (c) Immunohistochemical analysis of Htr2c-2A-iCre activity using a tdTomato reporter. Note high levels of tdTomato expression in the choroid plexus (CP). CC = corpus callosum. Similar results were obtained with at least 3 mice. Scale bar = 100 μm Source data.

Source data

Extended Data Fig. 3 EYFP-expressing LPBNHtr2c neurons are inhibited during sodium depletion.

(a) Cell-attached recordings taken from Htr2c-2A-iCre mice injected with AAV5-E1fa-DIO-EYFP. (b) Representative cell-attached recording of LPBNHtr2c neurons after 7 days of control diet, or 7 days of sodium-deficient diet. (c) Firing frequency of LPBNHtr2c neurons between control diet-fed mice (left) and sodium-deficient diet-fed mice (right). Two-tailed Mann-Whitney test. Control diet n = 21 cells, sodium-deficient diet n = 24 cells. Control diet 1.2 ± 0.3 Hz, Sodium-deficient diet 0.4 ± 0.2 Hz, P = 0.007. All data represented as mean ± s.e.m. **P < 0.01.

Extended Data Fig. 4 CNO injections in AAV-DIO-mCherry-injected controls do not affect food intake, water intake or 300 mM NaCl intake.

(a) Htr2c-2A-iCre mice injected with AAV2-hSyn-DIO-mCherry showed no changes in food intake, body weight or 300 mM NaCl intake. (b) No changes in water intake or 300 mM NaCl intake during sodium depletion. N = 5 mice. Water intake during sodium depletion: Two-Way Repeated Measures ANOVA: time (F(4, 16) = 20.52, P < 0.0001), treatment (F(1, 4) = 0.5421, P = 0.5024), interaction (F(4, 16) = 0.6448, P = 0.6385). 300 mM NaCl intake during sodium depletion: Two-Way Repeated Measures ANOVA: time (F(4,16) = 108.1, P < 0.0001), treatment (F(1, 4) = 0.1128, P = 0.7538), interaction (F(4, 16) = 0.3142, P = 0.8643). (c) No changes in food intake or body weight when using a control diet. N = 5 mice. Control diet food intake: Two-Way Repeated Measures ANOVA: time (F(3,12) = 223.9, P < 0.0001), treatment (F(1, 4) = 0.2222, P = 0.6619), interaction (F(3,12) = 2.689, P = 0.0924). All data represented as mean ± s.e.m.

Extended Data Fig. 5 LPBNHtr2c neuron activation can cause non-sodium related effects.

(a) Unilateral injection of AAV5-hSyn-DIO-hM3Dq into the LPBN of Htr2c-2A-iCre mice. (b) Activation of LPBNHtr2c neurons decreased water and 300 mM NaCl intake during dehydration in a two-bottle choice assay. N =11. Water intake during dehydration: Two-Way Repeated Measures ANOVA: time (F(4,40) = 67.06, P < 0.0001), treatment (F(1, 10) = 8.707, P = 0.0145), interaction (F(4, 40) = 6.329, P = 0.0005). Post hoc tests using the Bonferroni correction. 300 mM NaCl intake during dehydration: Two-Way Repeated Measures ANOVA: time (F(4,40) = 31.38, P < 0.0001), treatment (F(1, 10) = 17.76 P = 0.0018), interaction (F(4, 40) = 14.06 P = 0.0001). (c) Activation of LPBNHtr2c neurons decreased food during a fast refeeding regardless of sodium content. N = 11. Control diet food intake (left): Two-Way Repeated Measures ANOVA: time (F(3,30) = 111.0, P < 0.0001), treatment (F(1, 10) = 51.2, P < 0.0001), interaction (F(3, 30) = 32.00, P < 0.0001). Sodium-deficient diet food intake (right): Two-Way Repeated Measures ANOVA: time (F(3,30) = 50.59, P < 0.0001), treatment (F(1, 10) = 27.74, P = 0.0004), interaction (F(3, 30) = 18.81, P < 0.0001). All post hoc tests using the Bonferroni correction. All data represented as mean ± s.e.m. ****P < 0.0001.

Extended Data Fig. 6 LPBNHtr2c neurons colocalise with CGRP.

(a) Schematic of experiment. Unilateral injection of AAV5-EF1a-DIO -EYFP into the LPBN of Htr2c-2A-iCre mice to visualise LPBNHtr2c neurons (left). Colocalisation of EYFP labelled LPBNHtr2c neurons with CGRP (right). Cyan = EYFP, Magenta = CGRP, Blue = DAPI. Scale bar = 100 μm. (b) Estimation of colocalisation between LPBNHtr2c neurons and CGRP neurons. 3 slices taken from each mice. N = 7 mice.

Extended Data Fig. 7 Downstream circuitry of LPBNHtr2c neurons.

(a) Axonal projections visualised by unilateral injection of AAV5-EF1a-DIO-EYFP into the LPBN of Htr2c-2A-iCre mice. aca = anterior commissure (anterior part), acp = anterior commissure (posterior part), dBNST = bed nucleus of the stria terminalis (lateral division), fr = fasciculus retroflexus, ml = medial lemniscus, MnPO = median preoptic nucleus, opt = optic tract, PVH = paraventricular nucleus of the hypothalamus, scp = superior cerebellar peduncle, SPF = subparafascicular thalamus, st = stria terminalis, vBNST = bed nucleus of the stria terminalis (ventral part), VPPC = ventral posterior thalamus (parvicellular part). Scale bar = 400 μm. N = 7 mice. (b) Schematic of experiment. Bilateral injection of AAV2-DIO-hChR2 (H134R)-p2A-mCherry into the LPBN of Htr2c-2A-iCre mice (left). Voltage-clamp recordings taken at downstream sites (traces on the right). Blue rectangles indicate times of photostimulation.

Extended Data Fig. 8 Illustration showing the approximate spatial distribution of mCPP-responsive neurons within the LPBN.

C = central LPBN, D = dorsal LPBN, E = external LPBN, I = internal LPBN, KF = Kolliker’s Fuse, V = ventral LPBN.

Extended Data Fig. 9 Hyperpolarisation of LPBNHtr2c neurons is not dependent on Htr1b and is postsynaptic.

(a) Representative current clamp recording of an LPBNHtr2c neuron pre-treated with SB216641 (200 nM). Hyperpolarisation in response to mCPP persists in presence of SB216641. Arrows indicate time at which current steps were applied. (b) Voltage deflections in response to hyperpolarising currents from the same neuron, showing decreased input resistance in response to mCPP. Current steps in trace made in 10 pA increments from -50 pA to 0 pA. (c) Representative current clamp recording of an LPBNHtr2c neuron pre-treated with TTX (500 nM), kynurenic acid (KA, 1 mM) and picrotoxin (PTX, 50 μM), showing hyperpolarisation in response to mCPP. Arrows indicate time at which current steps were applied. (d) Voltage deflections in response to hyperpolarising currents from the same neuron, showing decreased input resistance in response to mCPP. Current steps in trace made in 10 pA increments from -50 pA to 0 pA. (e) Representative current clamp recording of an LPBNHtr2c neuron from a fasted mouse (18 hours), showing hyperpolarisation in response to mCPP. Arrows indicate time at which current steps were applied. (f) Voltage deflections in response to hyperpolarising currents from the same neuron, showing decreased input resistance in response to mCPP. Current steps in trace made in 10 pA increments from -50 pA to 0 pA. (g) Graph summarising change in membrane potential in response to mCPP under conditions tested. Neither pre-treatment with SB216641, Synaptic blockers nor recordings from fasted mice changed the magnitude of response to mCPP. mCPP n = 10 cells, SB216641 n = 5 cells, Synaptic block n = 3 cells, Fasted n = 8 cells. Data represented as mean ± s.e.m.

Extended Data Fig. 10 Serotoninergic projections to the LPBN.

(a) Schematic of experiment. Unilateral injection of Lumafluor retrobeads into the LPBN of wildtype mice to visualise upstream neurons. (b) Arrows indicate colocalised cells. Aq = aqueduct, DR = dorsal raphe. Scale bar = 200 μm. N = 6 mice (upper). MnR = median raphe. Scale bar = 100 μm. N = 6 mice (lower).

Supplementary information

Supplementary Information

Supplementary Table 1: Adjusted P values for post hoc comparisons.

Reporting Summary

Source data

Source Data Extended Data Fig. 2

Full gel picture for Extended Data Fig. 2b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, S., Williams, K.W., Liu, C. et al. A neural basis for tonic suppression of sodium appetite. Nat Neurosci 23, 423–432 (2020). https://doi.org/10.1038/s41593-019-0573-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0573-2

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