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:

Leptin-inhibited PBN neurons enhance responses to hypoglycemia in negative energy balance

This article has been updated

Abstract

Hypoglycemia initiates the counter-regulatory response (CRR), in which the sympathetic nervous system, glucagon and glucocorticoids restore glucose to appropriate concentrations. During starvation, low leptin levels restrain energy utilization, enhancing long-term survival. To ensure short-term survival during hypoglycemia in fasted animals, the CRR must overcome this energy-sparing program and nutrient depletion. Here we identify in mice a previously unrecognized role for leptin and a population of leptin-regulated neurons that modulate the CRR to meet these challenges. Hypoglycemia activates neurons of the parabrachial nucleus (PBN) that coexpress leptin receptor (LepRb) and cholecystokinin (CCK) (PBN LepRbCCK neurons), which project to the ventromedial hypothalamic nucleus. Leptin inhibits these cells, and Cckcre-mediated ablation of LepRb enhances the CRR. Inhibition of PBN LepRb cells blunts the CRR, whereas their activation mimics the CRR in a CCK-dependent manner. PBN LepRbCCK neurons are a crucial component of the CRR system and may be a therapeutic target in hypoglycemia.

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: VMH-projecting PBN LepRb neurons are activated by low glucose levels and are inhibited by leptin.
Figure 2: Deletion of LepRb from PBN CCK cells blunts IIH and augments the CRR to glucoprivation.
Figure 3: Enhanced CRR during hypoglycemic clamp in LepRbCck KO mice.
Figure 4: Inhibition of PBN LepRb neurons blunts the hyperglycemic response to glucoprivation.
Figure 5: Activation of PBN LepRb neurons mimics the CRR to glucoprivation.
Figure 6: CCK dependence of the hyperglycemic responses to activation of PBN LepRb neurons and glucoprivation.

Similar content being viewed by others

Change history

  • 10 December 2014

    In the version of this article initially published, the anteroposterior and dorsoventral stereotaxic injection coordinates in the Online Methods were transposed. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Levin, B.E., Dunn-Meynell, A.A. & Routh, V.H. CNS sensing and regulation of peripheral glucose levels. Int. Rev. Neurobiol. 51, 219–258 (2002).

    Article  CAS  Google Scholar 

  2. McCrimmon, R.J. & Sherwin, R.S. Hypoglycemia in type 1 diabetes. Diabetes 59, 2333–2339 (2010).

    Article  CAS  Google Scholar 

  3. Taborsky, G.J. Jr. Islets have a lot of nerve! Or do they? Cell Metab. 14, 5–6 (2011).

    Article  CAS  Google Scholar 

  4. Sanders, N.M. et al. Feeding and neuroendocrine responses after recurrent insulin-induced hypoglycemia. Physiol. Behav. 87, 700–706 (2006).

    Article  CAS  Google Scholar 

  5. Awoniyi, O., Rehman, R. & Dagogo-Jack, S. Hypoglycemia in patients with type 1 diabetes: epidemiology, pathogenesis, and prevention. Curr. Diab. Rep. 13, 669–678 (2013).

    Article  CAS  Google Scholar 

  6. Ahima, R.S. et al. Role of leptin in the neuroendocrine response to fasting. Nature 382, 250–252 (1996).

    Article  CAS  Google Scholar 

  7. Montague, C.T. et al. Congenital leptin deficiency is associated with severe early onset obesity in humans. Nature 387, 903–908 (1997).

    Article  CAS  Google Scholar 

  8. Farooqi, I.S. et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N. Engl. J. Med. 341, 879–884 (1999).

    Article  CAS  Google Scholar 

  9. Légrádi, G., Emerson, C.H., Ahima, R.S., Flier, J.S. & Lechan, R.M. Leptin prevents fasting-induced suppression of prothyrotropin-releasing hormone messenger ribonucleic acid in neurons of the hypothalamic paraventricular nucleus. Endocrinology 138, 2569–2576 (1997).

    Article  Google Scholar 

  10. Gautron, L. & Elmquist, J.K. Sixteen years and counting: an update on leptin in energy balance. J. Clin. Invest. 121, 2087–2093 (2011).

    Article  CAS  Google Scholar 

  11. Ahima, R.S., Saper, C.B., Flier, J.S. & Elmquist, J.K. Leptin regulation of neuroendocrine systems. Front. Neuroendocrinol. 21, 263–307 (2000).

    Article  CAS  Google Scholar 

  12. Morton, G.J., Cummings, D.E., Baskin, D.G., Barsh, G.S. & Schwartz, M.W. Central nervous system control of food intake and body weight. Nature 443, 289–295 (2006).

    Article  CAS  Google Scholar 

  13. Myers, M.G. Jr., Munzberg, H., Leinninger, G.M. & Leshan, R.L. The geometry of leptin action in the brain: more complicated than a simple ARC. Cell Metab. 9, 117–123 (2009).

    Article  CAS  Google Scholar 

  14. Friedman, J.M. Leptin at 14 y of age: an ongoing story. Am. J. Clin. Nutr. 89, 973S–979S (2009).

    Article  CAS  Google Scholar 

  15. McMinn, J.E. et al. Neuronal deletion of Lepr elicits diabesity in mice without affecting cold tolerance or fertility. Am. J. Physiol. Endocrinol. Metab. 289, E403–E411 (2005).

    Article  CAS  Google Scholar 

  16. de Luca, C. et al. Complete rescue of obesity, diabetes, and infertility in db/db mice by neuron-specific LEPR-B transgenes. J. Clin. Invest. 115, 3484–3493 (2005).

    Article  CAS  Google Scholar 

  17. Cohen, P. et al. Selective deletion of leptin receptor in neurons leads to obesity. J. Clin. Invest. 108, 1113–1121 (2001).

    Article  CAS  Google Scholar 

  18. Scott, M.M. et al. Leptin targets in the mouse brain. J. Comp. Neurol. 514, 518–532 (2009).

    Article  CAS  Google Scholar 

  19. Patterson, C.M., Leshan, R.L., Jones, J.C. & Myers, M.G. Jr. Molecular mapping of mouse brain regions innervated by leptin receptor–expressing cells. Brain Res. 1378, 18–28 (2011).

    Article  CAS  Google Scholar 

  20. Grill, H.J. Distributed neural control of energy balance: contributions from hindbrain and hypothalamus. Obesity (Silver Spring) 14 (suppl. 5), 216S–221S (2006).

    Article  Google Scholar 

  21. Ring, L.E. & Zeltser, L.M. Disruption of hypothalamic leptin signaling in mice leads to early-onset obesity, but physiological adaptations in mature animals stabilize adiposity levels. J. Clin. Invest. 120, 2931–2941 (2010).

    Article  CAS  Google Scholar 

  22. Vong, L. et al. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 71, 142–154 (2011).

    Article  CAS  Google Scholar 

  23. Leshan, R.L., Greenwald-Yarnell, M., Patterson, C.M., Gonzalez, I.E. & Myers, M.G. Jr. Leptin action through hypothalamic nitric oxide synthase-1–expressing neurons controls energy balance. Nat. Med. 18, 820–823 (2012).

    Article  CAS  Google Scholar 

  24. Hayes, M.R. et al. Endogenous leptin signaling in the caudal nucleus tractus solitarius and area postrema is required for energy balance regulation. Cell Metab. 11, 77–83 (2010).

    Article  CAS  Google Scholar 

  25. Scott, M.M., Williams, K.W., Rossi, J., Lee, C.E. & Elmquist, J.K. Leptin receptor expression in hindbrain Glp-1 neurons regulates food intake and energy balance in mice. J. Clin. Invest. 121, 2413–2421 (2011).

    Article  CAS  Google Scholar 

  26. Huo, L., Gamber, K.M., Grill, H.J. & Bjorbaek, C. Divergent leptin signaling in proglucagon neurons of the nucleus of the solitary tract in mice and rats. Endocrinology 149, 492–497 (2008).

    Article  CAS  Google Scholar 

  27. Huo, L., Maeng, L., Bjorbaek, C. & Grill, H.J. Leptin and the control of food intake: neurons in the nucleus of the solitary tract are activated by both gastric distension and Leptin. Endocrinology 148, 2189–2197 (2007).

    Article  CAS  Google Scholar 

  28. Garfield, A.S. et al. Neurochemical characterization of body weight–regulating leptin receptor neurons in the nucleus of the solitary tract. Endocrinology 153, 4600–4607 (2012).

    Article  CAS  Google Scholar 

  29. Opland, D. et al. Loss of neurotensin receptor-1 disrupts the control of the mesolimbic dopamine system by leptin and promotes hedonic feeding and obesity. Mol Metab. 2, 423–434 (2013).

    Article  CAS  Google Scholar 

  30. Levin, B.E., Magnan, C., Dunn-Meynell, A. & Le Foll, C. Metabolic sensing and the brain: who, what, where, and how? Endocrinology 152, 2552–2557 (2011).

    Article  CAS  Google Scholar 

  31. Levin, B.E., Becker, T.C., Eiki, J., Zhang, B.B. & Dunn-Meynell, A.A. Ventromedial hypothalamic glucokinase is an important mediator of the counterregulatory response to insulin-induced hypoglycemia. Diabetes 57, 1371–1379 (2008).

    Article  CAS  Google Scholar 

  32. Chan, O. et al. Increased GABAergic tone in the ventromedial hypothalamus contributes to suppression of counterregulatory responses after antecedent hypoglycemia. Diabetes 57, 1363–1370 (2008).

    Article  CAS  Google Scholar 

  33. Barnes, M.B., Lawson, M.A. & Beverly, J.L. Rate of fall in blood glucose and recurrent hypoglycemia affect glucose dynamics and noradrenergic activation in the ventromedial hypothalamus. Am. J. Physiol. Regul. Integr. Comp. Physiol. 301, R1815–R1820 (2011).

    Article  CAS  Google Scholar 

  34. Satoh, N. et al. Sympathetic activation of leptin via the ventromedial hypothalamus: leptin-induced increase in catecholamine secretion. Diabetes 48, 1787–1793 (1999).

    Article  CAS  Google Scholar 

  35. Takaki, A., Nagai, K., Takaki, S., Yanaihara, N. & Nakagawa, H. Satiety function of neurons containing a CCK-like substance in the dorsal parabrachial nucleus. Physiol. Behav. 48, 865–871 (1990).

    Article  CAS  Google Scholar 

  36. Yoshimatsu, H., Egawa, M. & Bray, G.A. Effects of cholecystokinin on sympathetic activity to interscapular brown adipose tissue. Brain Res. 597, 298–303 (1992).

    Article  CAS  Google Scholar 

  37. Le Lay, J. et al. CRTC2 (TORC2) contributes to the transcriptional response to fasting in the liver but is not required for the maintenance of glucose homeostasis. Cell Metab. 10, 55–62 (2009).

    Article  CAS  Google Scholar 

  38. Krashes, M.J. et al. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J. Clin. Invest. 121, 1424–1428 (2011).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  40. Wu, Q., Clark, M.S. & Palmiter, R.D. Deciphering a neuronal circuit that mediates appetite. Nature 483, 594–597 (2012).

    Article  CAS  Google Scholar 

  41. Leinninger, G.M. et al. Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance. Cell Metab. 14, 313–323 (2011).

    Article  CAS  Google Scholar 

  42. Song, P., Mabrouk, O.S., Hershey, N.D. & Kennedy, R.T. In vivo neurochemical monitoring using benzoyl chloride derivatization and liquid chromatography–mass spectrometry. Anal. Chem. 84, 412–419 (2012).

    Article  CAS  Google Scholar 

  43. Jiang, L., He, L. & Fountoulakis, M. Comparison of protein precipitation methods for sample preparation prior to proteomic analysis. J. Chromatogr. A 1023, 317–320 (2004).

    Article  CAS  Google Scholar 

  44. Kawamori, D. et al. Insulin signaling in alpha cells modulates glucagon secretion in vivo. Cell Metab. 9, 350–361 (2009).

    Article  CAS  Google Scholar 

  45. Jacobson, L., Ansari, T. & McGuinness, O.P. Counterregulatory deficits occur within 24 h of a single hypoglycemic episode in conscious, unrestrained, chronically cannulated mice. Am. J. Physiol. Endocrinol. Metab. 290, E678–E684 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank AstraZeneca Pharmaceuticals for the generous gift of leptin, B. Roth (University of North Carolina, Chapel Hill) for CNO and members of the labs of M.G.M. and L.K.H. for helpful discussions. Research support was provided by the Animal Phenotyping Core of the Michigan Diabetes Research Center (US National Institutes of Health (NIH) grant P30 DK020572) and the Michigan Nutrition and Obesity Research Center (P30 DK089503), the American Diabetes Association, the American Heart Association, the Marilyn H. Vincent Foundation and the NIH (DK098853) to M.G.M., NIH DK055267 and DK020595 to C.J.R., NIH DK046060 to R.T.K., NIH DK046409 to L.S. and the Wellcome Trust (098012) and Biotechnology and Biological Sciences Research Council (BB/K001418/1) to L.K.H.

Author information

Authors and Affiliations

Authors

Contributions

J.N.F. and C.M.P. produced the data in Figures 1,2,3,4,5 and Supplementary Figures 1,2,3,4,5,6,7,8,9,10, with the exception of the data in Figure 1e–i, which was produced by P.B.G. and L.S., and the measurements of catecholamines, which were performed by P.A.M., J.-M.T.W. and R.T.K. M.G. and M.R. helped produce Supplementary Figures 2, 5 and 10. J.C.J. aided with Figures 2,3,4 and with animal genotyping and husbandry. G.D., A.S.G. and L.K.H. performed the experiments in Figure 6. Adenoviral tracers were produced by A.K.S. and C.J.R. Experimental design, interpretation and manuscript preparation were led by M.G.M., L.K.H., J.N.F., C.M.P., A.S.G. and D.P.O.

Corresponding authors

Correspondence to Lora K Heisler or Martin G Myers Jr.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Leptin-responsive LepRb neurons in PBN.

LepRbeYFP mice were injected with IP (a) vehicle or (b) leptin (5 mg/kg) 2 hours prior to perfusion and immunostaining for pSTAT3 (purple) and GFP (eYFP; green). Representative of 2 similar experiments, each with n = 3 animals. The PBN of representative mice is shown. Scale bars = 100 μM. Inset- digital zoom of the indicated area. Arrowheads indicate examples of colocalized neurons; not all colocalized neurons are indicated.

Supplementary Figure 2 Activation of PBN LepRb cells by IIH and glucoprivation but not i.c.v. insulin.

(a,b) LepRbeYFP mice were injected with IP (a) vehicle or 2DG (250 mg/kg) or (b) vehicle or insulin (1 U/kg) 2 hours prior to perfusion and immunostaining for cFos (purple) and GFP (eYFP; green) *p<0.001, {t(13) = −4.371}. (c,d) LepRbeYFP mice were injected with ICV (c) *p = 0.008, {t(12) = −3.19} vehicle or 2DG (1µg) *p<0.001, {t(9) = −6.01} or (d) vehicle or insulin (300mU) 2 hours prior to perfusion and immunostaining for cFos (purple) and GFP (eYFP; green). Colocalized cFos+eYFP and total eYFP neurons were quantified; data are presented as %colocalized/total eYFP. All studies: n = 5–8 animals analyzed by one tailed t-test; each panel representative of two separate studies.

Supplementary Figure 3 Similar pattern of cFos IR in PBN LepRb cells in response to i.p. insulin and 2DG.

LepRbeYFP mice were injected with IP (a) vehicle, (b) insulin (1 U/kg), or or 2DG (250 mg/kg) 2 hours prior to perfusion and immunostaining for cFos (purple) and GFP (eYFP; green). The PBN of representative mice is shown. Scale bars= 100 μM. n = 9 animals per group; representative of two separate studies. Inset- digital zoom of the indicated area. Arrowheads indicate examples of colocalized neurons; not all colocalized neurons are indicated.

Supplementary Figure 4 Leptin blunts fasting-induced enhancement of the CRR.

Male C57Bl6/J mice were allowed to feed ad libitum (Control), were fasted for 12 hours (Fast), or were injected with leptin (5 mg/kg, IP) and fasted for 12 hours (Fast+Leptin) before being injected with 2DG (500 mg/kg, IP). (a) Blood glucose was monitored at the indicated times; p = 0.013 (60 min), <0.001 (90 min), 0.008 (120 min) vs control and p = 0.003 (90 min), 0.008 (120 min) vs Fast + Leptin {F(12,26) = 4.61} by two way repeated measures ANOVA with Fisher LSD post hoc test. N = 8–9 animals per condition; data are plotted as mean +/− SEM. Letters indicate significant differences relative to other conditions by ANOVA with Fishers post-hoc test. (b) Area under the curve (AUC) for the data graphed in (a); *p<0.001 vs control and *0.009 vs Fast + Leptin {F(2,26) = 15.15}by one way ANOVA with Fisher LSD post hoc test.

Supplementary Figure 5 Induction of cFos in PBN CCK neurons by glucoprivation.

(a,b) CCKeYFP mice were injected with IP (a) vehicle or (b) 2DG (250 mg/kg, IP) 2 hours prior to perfusion and immunostaining for cFos (purple) and GFP (eYFP; green). The PBN of representative mice is shown. N=11 animals per condition; representative of two similar studies. Scale bars= 100 μM. Inset- digital zoom of the indicated area. Arrowheads indicate examples of colocalized neurons; not all colocalized neurons are indicated. (c) Quantification of the data. Colocalized cFos+eYFP and total eYFP neurons were quantified; data are presented as %colocalized/total eYFP; *p = 0.017, {t(16) = −2.32}. n = 7–11 animals per condition *p<0.05 by one tailed t-test; representative of two separate studies.

Supplementary Figure 6 Reduced leptin-stimulated pSTAT3 IR in PBN and EW nuclei of LepRbCck KO mice.

Quantitation of pSTAT3-IR cells in the indicated hindbrain nuclei of Control and LepRbCckKO mice following treatment with leptin (IP, 5 mg/kg) 2 hours prior to perfusion. N = 3–4 animals per condition; data are plotted as mean +/− SEM. NTS- nucleus tractus solitarius; PBN- parabrachial nucleus; PAG- periaqueductal grey; DR- dorsal raphe; EW- Edinger-Westphal; LR- linear raphe. EW: t(5) = 4.01, *p = 0.05. PBN: t(5) = 8.28, *p<0.001 by one tailed t-test.

Supplementary Figure 7 Energy balance and glucose homeostasis in LepRbCck KO mice.

(a) Weekly body weight for male Control and LepRbCckKO mice. (b) GTT (blood glucose values at the indicated times following an IP injection of 3 g/kg) in 15-week old male Control and LepRbCckKO mice. Hormone levels at 14 weeks of age in male Control and LepRbCckKO mice: (c) leptin, (d) insulin, (e) corticosterone. N = 12–18 animals per condition; p = NS for all comparisons; all data are plotted as mean +/− SEM.

Supplementary Figure 8 Fasting fails to enhance the CRR to glucoprivation in LepRbCck KO mice.

Male Control and LepRbCckKO mice were fasted for 4 (a,b) or 24 (c,d) hours prior to injection with 2DG (250 mg/kg, IP); blood glucose was monitored for 180 minutes afterwards (a,c); *p<0.001 (90 min), <0.001 (12 0min), {F(6,20) = 19.43} by two way repeated measures ANOVA with Fisher LSD post hoc test. (b,d) Quantification of area under the curve (AUC) from the data in (a,c) respectively; *p = 0.012, {t(19) = −2.79} by two tailed t-test. N = 9–13 animals per condition; all data are plotted as mean +/− SEM.

Supplementary Figure 9 Leptin action via PBNCck neurons does not alter glycemic responses to restraint stress.

(a) Leprcre mice were crossed onto the Rosa26-eYFP background to generate LepRbeYFP mice expressing eYFP in LepRb neurons. (b,c) LepRbeYFP mice were handled briefly (b) or subjected to restraint stress in a conical tube for 30 minutes (c) and then were perfused 2 hours prior to immunostaining for cFos (purple) and GFP (eYFP; green) in one experiment. The PBN of representative mice is shown. Inset- digital zoom of the indicated area. Arrowheads indicate examples of colocalized neurons. (d) Cckcre mice were crossed onto the Leprflox background to generate LepRbCckKO mice. (e,f) Control and LepRbCckKOeYFP mice were subjected to restraint stress for 30 minutes and blood glucose was monitored for 120 minutes (e). (f) Area under the curve (AUC) is plotted for the data in (e). N = 2–4 animals per condition; p = NS for all comparisons; all data are plotted as mean +/− SEM.

Supplementary Figure 10 Induction of cFos in PBN and VMH neurons after DREADD-mediated activation of PBN LepRb neurons.

The cre-inducible AAV-hM3Dq was injected bilaterally into the PBN of male Leprcre mice. Animals were injected with Vehicle (a,c) or CNO (b,d; 0.3 mg/kg, IP) and monitored for 120 minutes (see Figure 6). At the end of the experiment, animals were perfused and brain sections were stained for cFos (green; panels a-d) and/or dsRed (red; panels a-b). Representative images are shown for the PBN (a,b) and VMH (c,d) in one experiment. (e) Quantification of cFos in the VMH of Vehicle and CNO treated animals. N = 9–11 animals per condition; *p<0.001, {t(18) = −6.83} by one-tailed t-test; all data are plotted as mean +/− SEM.

Supplementary Figure 11 Working model of PBN LepRb neurons.

Our data suggest that PBN LepRb expressing neurons are excited by hypoglycemia, inhibited by leptin, release CCK in the VMH to drive counterregulatory responses. Our data indicates the importance of this pathway in modulating counter-regulatory responses during fasting conditions to promote survival of the individual.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 (PDF 1397 kb)

Supplementary Methods Checklist

(PDF 421 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Flak, J., Patterson, C., Garfield, A. et al. Leptin-inhibited PBN neurons enhance responses to hypoglycemia in negative energy balance. Nat Neurosci 17, 1744–1750 (2014). https://doi.org/10.1038/nn.3861

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3861

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