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.

  • Brief Communication
  • Published:

HSD2 neurons in the hindbrain drive sodium appetite

Abstract

Sodium-depleted animals develop an appetite for aversive concentrations of sodium. Here we show that chemogenetic activation of aldosterone-sensitive neurons that express 11β-hydroxysteroid dehydrogenase type 2 (HSD2) in the nucleus of the solitary tract is sufficient to drive consumption of sodium-containing solutions in mice, independently of thirst or hunger. These HSD2-positive neurons are necessary for full expression of sodium appetite and have distinct downstream targets that are activated during sodium depletion.

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

Figure 1: HSD2 neurons generate a sodium appetite independently of thirst.
Figure 2: The appetite induced by HSD2 neurons is specific for sodium.
Figure 3: Efferent projections from HSD2 neurons.

Similar content being viewed by others

References

  1. Luquet, S., Perez, F.A., Hnasko, T.S. & Palmiter, R.D. Science 310, 683–685 (2005).

    Article  CAS  Google Scholar 

  2. Oka, Y., Ye, M. & Zuker, C.S. Nature 520, 349–352 (2015).

    Article  CAS  Google Scholar 

  3. Aponte, Y., Atasoy, D. & Sternson, S.M. Nat. Neurosci. 14, 351–355 (2011).

    Article  CAS  Google Scholar 

  4. Geerling, J.C. & Loewy, A.D. Exp. Physiol. 93, 177–209 (2008).

    Article  CAS  Google Scholar 

  5. Formenti, S. et al. Am. J. Physiol. Regul. Integr. Comp. Physiol. 304, R252–R259 (2013).

    Article  CAS  Google Scholar 

  6. Geerling, J.C., Kawata, M. & Loewy, A.D. J. Comp. Neurol. 494, 515–527 (2006).

    Article  Google Scholar 

  7. Chapman, K., Holmes, M. & Seckl, J. Physiol. Rev. 93, 1139–1206 (2013).

    Article  CAS  Google Scholar 

  8. Rogan, S.C. & Roth, B.L. Pharmacol. Rev. 63, 291–315 (2011).

    Article  CAS  Google Scholar 

  9. Sakai, R.R., Frankmann, S.P., Fine, W.B. & Epstein, A.N. Behav. Neurosci. 103, 186–192 (1989).

    Article  CAS  Google Scholar 

  10. Antunes-Rodrigues, J., De Castro, M., Elias, L.L.K., Valença, M.M. & McCann, S.M. Physiol. Rev. 84, 169–208 (2004).

    Article  CAS  Google Scholar 

  11. Dadam, F.M. et al. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306, R175–R184 (2014).

    Article  CAS  Google Scholar 

  12. Kim, J.C. et al. Neuron 63, 305–315 (2009).

    Article  CAS  Google Scholar 

  13. Evans, L.C. et al. Circulation 133, 1360–1370 (2016).

    Article  CAS  Google Scholar 

  14. Breslin, P.A., Spector, A.C. & Grill, H.J. Am. J. Physiol. 264, R319–R323 (1993).

    CAS  PubMed  Google Scholar 

  15. Krashes, M.J. et al. J. Clin. Invest. 121, 1424–1428 (2011).

    Article  CAS  Google Scholar 

  16. Geerling, J.C. & Loewy, A.D. J. Comp. Neurol. 498, 223–250 (2006).

    Article  Google Scholar 

  17. Geerling, J.C. et al. Brain Res. 1375, 19–27 (2011).

    Article  CAS  Google Scholar 

  18. Menani, J.V., De Luca, L.A. Jr. & Johnson, A.K. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306, R201–R210 (2014).

    Article  CAS  Google Scholar 

  19. Betley, J.N., Cao, Z.F.H., Ritola, K.D. & Sternson, S.M. Cell 155, 1337–1350 (2013).

    Article  CAS  Google Scholar 

  20. Nation, H.L., Nicoleau, M., Kinsman, B.J., Browning, K.N. & Stocker, S.D. J. Neurophysiol. 115, 3123–3129 (2016).

    Article  CAS  Google Scholar 

  21. Gore, B.B., Soden, M.E. & Zweifel, L.S. Curr. Protoc. Neurosci. 4, 4.35.1–4.35.20 (2013).

    Google Scholar 

  22. Liedtke, W.B. et al. Proc. Natl. Acad. Sci. USA 108, 12509–12514 (2011).

    Article  CAS  Google Scholar 

  23. Geerling, J.C. et al. Am. J. Physiol. Regul. Integr. Comp. Physiol. 310, R41–R54 (2016).

    Article  Google Scholar 

  24. De Solis, A.J., Baquero, A.F., Bennett, C.M., Grove, K.L. & Zeltser, L.M. Mol. Metab. 5, 189–209 (2016).

    Google Scholar 

  25. Ma, C.-W. et al. J. Comp. Neurol. 521, 612–625 (2013).

    Article  CAS  Google Scholar 

  26. Enjin, A. et al. J. Comp. Neurol. 518, 2284–2304 (2010).

    Article  CAS  Google Scholar 

  27. Bochorishvili, G., Stornetta, R.L., Coates, M.B. & Guyenet, P.G. J. Comp. Neurol. 520, 1047–1061 (2012).

    Article  CAS  Google Scholar 

  28. Rowland, N.E., Farnbauch, L.J. & Crews, E.C. Physiol. Behav. 80, 629–635 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Kafer and M. Chiang for help generating and maintaining the Hsd11b2Cre mouse line. We thank H. King for assistance with behavior and histology. We thank the Palmiter laboratory and J. Schulkin for discussion and critiques. This work was supported by the National Science Foundation Graduate Research Fellowship under grant no. DGE-1256082 (B.C.J.). Any opinions, findings, conclusions or recommendations expressed in this material are those of the authors and do not necessarily reflect the views of the National Science Foundation.

Author information

Authors and Affiliations

Authors

Contributions

B.C.J. designed experiments under the guidance of R.D.P.; B.C.J. performed and analyzed experiments. B.C.J. and R.D.P. wrote the manuscript.

Corresponding authors

Correspondence to Brooke C Jarvie or Richard D Palmiter.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Function of HSD2 and generation of Hsd11b2Cre mice.

(a) HSD2 converts corticosterone to an inactive form that can no longer compete with aldosterone for the mineralocortioid receptor. (b) Diagram showing: top, the Hsd11b2 gene (5 exons spanning ~5.5 kb with coding region in dark blue). Middle, the targeting vector; Bottom, Hsd11b2 locus after recombination and removal of the frt-flanked SV-40 NeoR selection gene. Some key restriction enzyme sites used for cloning are shown. Diagram is not to scale.

Supplementary Figure 2 CNO activates virally targeted HSD2 neurons in the NTS.

(a) Representative sections from mCherry or hM3Dq:mCherry-transduced Hsd11b2Cre animals following i.p. injection of CNO (1.0 mg/kg) and subsequent immunohistochemical staining for Fos. Scale bar, 200 μm. (b, c) Quantification of the percentage of mCherry-expressing neurons in the NTS co-expressing Fos in (b) male (n = 7 mice per group, two-tailed, unpaired t-test, P = 5.17E-10) and (c) female (n = 3 and 4 mice; two-tailed, unpaired t-test, P = 3.559E-06) Hsd11b2Cre mice. AP, area postrema. cc, central canal.

Supplementary Figure 3 Effects of hM4Di-mediated inhibition of HSD2 neurons.

(a) There was no change in sodium intake following sodium-depletion in Hsd11b2Cre control mice expressing mCherry following injections of either saline or CNO (1mg/kg) (Repeated measure ANOVA, P = 0.382). (b) There was no change in water intake during the first 2-h after NaCl was returned to sodium depleted Hsd11b2Cre mice expressing either mCherry or hM4Di:mCherry (paired salt intake data from from panel a and Fig. 1f) (Repeated measure ANOVA, P = 0.589). (c) CNO administration in sodium-depleted mice decreased Fos expression in the NTS of mice that expressed hM4Di:mCherry as compared to mCherry (unpaired, two-tailed t-test, P = 0.031). (d) Representative image showing mCherry fluorescence in HSD2 cells at the rostral-caudal level where Fos is shown in the panel e. (e) Representative image showing that CNO administration decreased Fos-immunoreactivity in the NTS of hM4Di:mCherry mice following sodium depletion, but no in mCherry controls. It was difficult to quantify HSD2 cell bodies in the hM4Di:mCherry-expressing mice, so Fos is not shown as a percentage of HSD2 neurons but rather represented as overall expression in the NTS. Scale bar, 100 μm. cc, central canal.

Supplementary Figure 4 Chronic inactivation of HSD2 neurons is fatal.

HSD2 neurons were targeted in the NTS with AAV-DIO-GFP:TetTox in a mix of male (n = 2) and female (n = 3) Hsd11b2Cre mice. HSD2 neurons were transduced with AAV-DIO-YFP in control mice (male, n = 1; female, n = 5). (a) Mice became ungroomed (data not shown) and dropped to 80% of their pre-surgery body weight, at which point they were euthanized. On day 9, 3/5 mice were removed from the study (shown on graph), and the remaining 2/2 dropped to 80% and were euthanized on day 10 (Day 9, ** P = 0.009). (b) Overnight food (Day 7 * P = 0.039, day 8 **** P < 0.0001)(c) and water intake (Day 7 * P = 0.028, day 8 ** P = 0.008) both dropped the night before mice started to rapidly lose weight. Repeated measure ANOVA with post-hoc Sidak’s multiple comparisons were used.

Supplementary Figure 5 Mice increased KCl consumption following sodium depletion.

Male wild-type mice increased their intake of 0.5 M KCl but not CaCl2 following furosemide-induced sodium depletion (Repeated measures ANOVA, Sidak post hoc. KCl ** P = 0.007, CaCl2 P = 0.999). Some of these mice had undergone a bout of sodium depletion (with at least a week recovery time) prior to this test.

Supplementary Figure 6 Activation of HSD2 neurons does not alter water intake.

Water intake (4 h) expressed as a percentage of baseline intake from two-bottle choice assays in response to CNO (1 mg/kg) administration or the following night (Recovery). (a, b) Water intake in male mice from taste assays in Fig. 2b or paired with food intake from Fig. 2d (two-way ANOVA. CNO Stim, P = 0.6930. Recovery, P = 0.3293) Water intake is not shown from the sucrose taste test because mice predominantly drank sucrose instead of water, and water was not measured during the 3-bottle assay in Fig. 2c. (c, d) Water intake in female mice during the two-bottle choice tests shown in Fig. 1c, or paired with food intake from Fig. 2d (two-way ANOVA. CNO Stim, P = 0.1663. Recovery, P = 0.2376).

Supplementary Figure 7 Differential Fos induction in the BNSTv.

(a) Representative sections of immunoreactivity for Fos in the BNSTv of male mice with mCherry expressed in terminals of HSD2 neurons originating in the NTS. Scale bar, 60 μm. (b) Schematic showing anatomy of the ventral BNST. (c) A circle was drawn around fiber terminals, and Fos was counted within the circle (Fig. 3d), and (c) throughout the rest of the BNSTv. No changes in the number of Fos-immunoreactive cells were detected that were not directly under the fibers (multiple, two-tailed t-tests with Holm-Sidak post hoc. Hsd11b2, P = 0.652. Na-dep, P = 0.626). aco, anterior commissure; am, anteromedial; al, anterolateral; fu, fusiform; ps, parastrial nucleus.

Supplementary Figure 8 Fos induction in the PBN.

(a) Representative images showing location of Foxp2 and Fos immunoreactivity in the mPBcl of the lateral parabrachial nucleus in response to sodium-depletion, or mCherry and hM3Dq:mCherry-expressing Hsd11b2Cre mice in response to CNO. Scale bar, 60 μm. (b) Schematic showing relative location of neurons that co-express Foxp2 and Fos (yellow). (c) Quantification of neurons co-expressing Foxp2 and Fos in the rest of the central lateral (cl) and dorsal lateral (dl) region of the parabrachial nucleus in the same sections counted for Fig 3b (multiple, two-tailed t-tests with Holm-Sidak post hoc. Hsd11b2, P = 0.9510. Na-dep, P = 0.312). scp, superior cerebellar peduncle; vl, ventral lateral; elo, external lateral; MPB, medial parabrachial nucleus.

Supplementary Figure 9 Fos induction in the pre-LC and surrounding region.

(a) Representative images showing location of Foxp2 and Fos immunoreactivity in the pre-LC (pLC) and the region of Barrington’s nucleus (B, also see panel b) in response to sodium depletion, or in response to CNO administration in mCherry and hM3Dq:mCherry-expressing Hsd11b2Cre mice. Scale bar, 60 μm. (b) Schematic showing relative location of Foxp2 and Fos neurons (green). (c) Quantification of neurons co-expressing Foxp2 and Fos in the region of Barrington’s nucleus (multiple, two-tailed t-tests with Holm-Sidak post hoc. Hsd11b2, P = 0.0003. Na-dep, P = 0.0018). See Fig. 3c for pLC quantification. LC, locus coeruleus; MeV, midbrain trigeminal nucleus; scp, superior cerebellar peduncle.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1 and 2 (PDF 5749 kb)

Supplementary Methods Checklist (PDF 468 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jarvie, B., Palmiter, R. HSD2 neurons in the hindbrain drive sodium appetite. Nat Neurosci 20, 167–169 (2017). https://doi.org/10.1038/nn.4451

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4451

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