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 rapidly acting glutamatergic ARC→PVH satiety circuit postsynaptically regulated by α-MSH

A Corrigendum to this article was published on 26 July 2017

This article has been updated

Abstract

Arcuate nucleus (ARC) neurons sense the fed or fasted state and regulate hunger. Agouti-related protein (AgRP) neurons in the ARC (ARCAgRP neurons) are stimulated by fasting and, once activated, they rapidly (within minutes) drive hunger. Pro-opiomelanocortin (ARCPOMC) neurons are viewed as the counterpoint to ARCAgRP neurons. They are regulated in an opposite fashion and decrease hunger. However, unlike ARCAgRP neurons, ARCPOMC neurons are extremely slow in affecting hunger (many hours). Thus, a temporally analogous, rapid ARC satiety pathway does not exist or is presently unidentified. Here we show that glutamate-releasing ARC neurons expressing oxytocin receptor, unlike ARCPOMC neurons, rapidly cause satiety when chemo- or optogenetically manipulated. These glutamatergic ARC projections synaptically converge with GABAergic ARCAgRP projections on melanocortin-4 receptor (MC4R)-expressing satiety neurons in the paraventricular hypothalamus (PVHMC4R neurons). Transmission across the ARCGlutamatergic→PVHMC4R synapse is potentiated by the ARCPOMC neuron-derived MC4R agonist, α-melanocyte stimulating hormone (α-MSH). This excitatory ARC→PVH satiety circuit, and its modulation by α-MSH, provides insight into regulation of hunger and satiety.

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: Regulation of feeding by ARCPOMC versus ARCVglut2 neurons.
Figure 2: Effects of fasting on ARCVglut2 neurons and their inhibition by ARCAgRP neuron afferents.
Figure 3: ARCVglut2→PVH projections, unlike ARCPOMC→PVH projections, are effective in glutamatergic transmission.
Figure 4: RNA-seq analysis of ARCVglut2 neurons.
Figure 5: Oxtr expression marks ARCGlutamatergic neurons that rapidly promote satiety.
Figure 6: ARCGlutamatergic and ARCOxtr neurons preferentially engage PVHMC4R neurons.
Figure 7: α-MSH postsynaptically increases excitatory input onto PVHMC4R neurons.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 20 February 2017

    In the version of this article initially published, α-MSH was referred to at the beginning of the fourth paragraph of the introduction as an MC4R antagonist. This should have read agonist. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Langlet, F. Tanycytes: a gateway to the metabolic hypothalamus. J. Neuroendocrinol. 26, 753–760 (2014).

    CAS  PubMed  Google Scholar 

  2. Rodríguez, E.M., Blázquez, J.L. & Guerra, M. The design of barriers in the hypothalamus allows the median eminence and the arcuate nucleus to enjoy private milieus: the former opens to the portal blood and the latter to the cerebrospinal fluid. Peptides 31, 757–776 (2010).

    PubMed  Google Scholar 

  3. Xu, A.W. et al. Effects of hypothalamic neurodegeneration on energy balance. PLoS Biol. 3, e415 (2005).

    PubMed  PubMed Central  Google Scholar 

  4. Zhan, C. et al. Acute and long-term suppression of feeding behavior by POMC neurons in the brainstem and hypothalamus, respectively. J. Neurosci. 33, 3624–3632 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Krude, H. et al. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat. Genet. 19, 155–157 (1998).

    CAS  PubMed  Google Scholar 

  6. Yaswen, L., Diehl, N., Brennan, M.B. & Hochgeschwender, U. Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nat. Med. 5, 1066–1070 (1999).

    CAS  PubMed  Google Scholar 

  7. Huszar, D. et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88, 131–141 (1997).

    CAS  PubMed  Google Scholar 

  8. Balthasar, N. et al. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 123, 493–505 (2005).

    CAS  PubMed  Google Scholar 

  9. Vaisse, C., Clement, K., Guy-Grand, B. & Froguel, P. A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nat. Genet. 20, 113–114 (1998).

    CAS  PubMed  Google Scholar 

  10. Yeo, G.S. et al. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat. Genet. 20, 111–112 (1998).

    CAS  PubMed  Google Scholar 

  11. Luquet, S., Perez, F.A., Hnasko, T.S. & Palmiter, R.D. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310, 683–685 (2005).

    CAS  PubMed  Google Scholar 

  12. Aponte, Y., Atasoy, D. & Sternson, S.M. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat. Neurosci. 14, 351–355 (2011).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Myers, M.G. Jr. & Olson, D.P. Central nervous system control of metabolism. Nature 491, 357–363 (2012).

    CAS  PubMed  Google Scholar 

  15. Morton, G.J., Meek, T.H. & Schwartz, M.W. Neurobiology of food intake in health and disease. Nat. Rev. Neurosci. 15, 367–378 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gautron, L., Elmquist, J.K. & Williams, K.W. Neural control of energy balance: translating circuits to therapies. Cell 161, 133–145 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Koch, M. et al. Hypothalamic POMC neurons promote cannabinoid-induced feeding. Nature 519, 45–50 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Krashes, M.J., Shah, B.P., Koda, S. & Lowell, B.B. Rapid versus delayed stimulation of feeding by the endogenously released AgRP neuron mediators GABA, NPY, and AgRP. Cell Metab. 18, 588–595 (2013).

    CAS  PubMed  Google Scholar 

  19. Bagnol, D. et al. Anatomy of an endogenous antagonist: relationship between Agouti-related protein and proopiomelanocortin in brain. J. Neurosci. 19, RC26 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Cowley, M.A. et al. Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron 24, 155–163 (1999).

    CAS  PubMed  Google Scholar 

  21. Giraudo, S.Q., Billington, C.J. & Levine, A.S. Feeding effects of hypothalamic injection of melanocortin 4 receptor ligands. Brain Res. 809, 302–306 (1998).

    CAS  PubMed  Google Scholar 

  22. Shah, B.P. et al. MC4R-expressing glutamatergic neurons in the paraventricular hypothalamus regulate feeding and are synaptically connected to the parabrachial nucleus. Proc. Natl. Acad. Sci. USA 111, 13193–13198 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Atasoy, D., Betley, J.N., Su, H.H. & Sternson, S.M. Deconstruction of a neural circuit for hunger. Nature 488, 172–177 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Krashes, M.J. et al. An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature 507, 238–242 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Atasoy, D. et al. A genetically specified connectomics approach applied to long-range feeding regulatory circuits. Nat. Neurosci. 17, 1830–1839 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Branco, T. et al. Near-perfect synaptic integration by Nav1.7 in hypothalamic neurons regulates body weight. Cell 165, 1749–1761 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Balthasar, N. et al. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 42, 983–991 (2004).

    CAS  PubMed  Google Scholar 

  30. Atasoy, D., Aponte, Y., Su, H.H. & Sternson, S.M. A FLEX switch targets Channelrhodopsin-2 to multiple cell types for imaging and long-range circuit mapping. J. Neurosci. 28, 7025–7030 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Petreanu, L., Huber, D., Sobczyk, A. & Svoboda, K. Channelrhodopsin-2-assisted circuit mapping of long-range callosal projections. Nat. Neurosci. 10, 663–668 (2007).

    CAS  PubMed  Google Scholar 

  32. Hahn, T.M., Breininger, J.F., Baskin, D.G. & Schwartz, M.W. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat. Neurosci. 1, 271–272 (1998).

    CAS  PubMed  Google Scholar 

  33. Jarvie, B.C. & Hentges, S.T. Expression of GABAergic and glutamatergic phenotypic markers in hypothalamic proopiomelanocortin neurons. J. Comp. Neurol. 520, 3863–3876 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wittmann, G., Hrabovszky, E. & Lechan, R.M. Distinct glutamatergic and GABAergic subsets of hypothalamic pro-opiomelanocortin neurons revealed by in situ hybridization in male rats and mice. J. Comp. Neurol. 521, 3287–3302 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Cravo, R.M. et al. Characterization of Kiss1 neurons using transgenic mouse models. Neuroscience 173, 37–56 (2011).

    CAS  PubMed  Google Scholar 

  36. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    CAS  PubMed  Google Scholar 

  37. Usoskin, D. et al. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat. Neurosci. 18, 145–153 (2015).

    CAS  PubMed  Google Scholar 

  38. Südhof, T.C. Neurotransmitter release: the last millisecond in the life of a synaptic vesicle. Neuron 80, 675–690 (2013).

    PubMed  Google Scholar 

  39. Blakely, R.D. & Edwards, R.H. Vesicular and plasma membrane transporters for neurotransmitters. Cold Spring Harb. Perspect. Biol. 4, a005595 (2012).

    PubMed  PubMed Central  Google Scholar 

  40. Maejima, Y. et al. Oxytocinergic circuit from paraventricular and supraoptic nuclei to arcuate POMC neurons in hypothalamus. FEBS Lett. 588, 4404–4412 (2014).

    CAS  PubMed  Google Scholar 

  41. Nakajima, M., Görlich, A. & Heintz, N. Oxytocin modulates female sociosexual behavior through a specific class of prefrontal cortical interneurons. Cell 159, 295–305 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lim, B.K., Huang, K.W., Grueter, B.A., Rothwell, P.E. & Malenka, R.C. Anhedonia requires MC4R-mediated synaptic adaptations in nucleus accumbens. Nature 487, 183–189 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Shen, Y., Fu, W.Y., Cheng, E.Y., Fu, A.K. & Ip, N.Y. Melanocortin-4 receptor regulates hippocampal synaptic plasticity through a protein kinase A-dependent mechanism. J. Neurosci. 33, 464–472 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Blevins, J.E. et al. Chronic oxytocin administration inhibits food intake, increases energy expenditure, and produces weight loss in fructose-fed obese rhesus monkeys. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R431–R438 (2015).

    CAS  PubMed  Google Scholar 

  45. Lawson, E.A. et al. Oxytocin reduces caloric intake in men. Obesity (Silver Spring) 23, 950–956 (2015).

    CAS  Google Scholar 

  46. Blevins, J.E. & Baskin, D.G. Translational and therapeutic potential of oxytocin as an anti-obesity strategy: Insights from rodents, nonhuman primates and humans. Physiol. Behav. 152B, 438–449 (2015).

    Google Scholar 

  47. van den Pol, A.N. et al. Neuromedin B and gastrin-releasing peptide excite arcuate nucleus neuropeptide Y neurons in a novel transgenic mouse expressing strong Renilla green fluorescent protein in NPY neurons. J. Neurosci. 29, 4622–4639 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Tong, Q., Ye, C.P., Jones, J.E., Elmquist, J.K. & Lowell, B.B. Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat. Neurosci. 11, 998–1000 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    CAS  PubMed  Google Scholar 

  50. Farley, F.W., Soriano, P., Steffen, L.S. & Dymecki, S.M. Widespread recombinase expression using FLPeR (flipper) mice. Genesis 28, 106–110 (2000).

    CAS  PubMed  Google Scholar 

  51. Padilla, S.L., Reef, D. & Zeltser, L.M. Defining POMC neurons using transgenic reagents: impact of transient Pomc expression in diverse immature neuronal populations. Endocrinology 153, 1219–1231 (2012).

    CAS  PubMed  Google Scholar 

  52. Jovanovic, Z., Tung, Y.C., Lam, B.Y., O'Rahilly, S. & Yeo, G.S. Identification of the global transcriptomic response of the hypothalamic arcuate nucleus to fasting and leptin. J. Neuroendocrinol. 22, 915–925 (2010).

    CAS  PubMed  Google Scholar 

  53. Henry, F.E., Sugino, K., Tozer, A., Branco, T. & Sternson, S.M. Cell type-specific transcriptomics of hypothalamic energy-sensing neuron responses to weight-loss. Elife 4, e09800 (2015).

    PubMed Central  Google Scholar 

  54. Burbach, J.P. Neuropeptides from concept to online database www.neuropeptides.nl. Eur. J. Pharmacol. 626, 27–48 (2010).

    CAS  PubMed  Google Scholar 

  55. Kong, D. et al. GABAergic RIP-Cre neurons in the arcuate nucleus selectively regulate energy expenditure. Cell 151, 645–657 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of B.B.L.'s laboratory for discussions. This research was funded by the following NIH grants to B.B.L.: R01 DK075632, R01 DK096010, R01 DK089044, R01 DK111401, P30 DK046200, P30 DK057521; to J.M.R.: F32 DK103387; to J.N.C.: AHA Postdoctoral Fellowship 14POST20100011. Y.M.-C. was supported by a Charles A. King postdoctoral fellowship. Y.L. was supported by an EMBO postdoctoral fellowship.

Author information

Authors and Affiliations

Authors

Contributions

H.F. and B.B.L. designed the experiments and analyzed data. J.N.C. performed the single-cell RNA-seq experiments with help from Y.M.-C. H.F. and A.M.J.V. conducted the behavioral and histological studies with help from J.M.R. and Z.Y. H.F. performed the electrophysiological experiments with help from J.C.M. and Y.L. J.X. generated Pomc-IRES-cre mice. B.P.S. generated PomcloxP mice. H.F. and B.B.L. wrote the manuscript with comments from all of the authors.

Corresponding author

Correspondence to Bradford B Lowell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Validation of hM3Dq expression and function in ARCPOMC neurons; generation and validation of Pomc-IRES-Cre mice.

a, Experimental schematics. b, Expression of AAV-DIO-hM3Dq-mCherry in ARCPOMC neurons in Pomc-Cre mice determined by mCherry immunolabeling (magenta). c, Representative trace of cell-attached recordings from a hM3Dq-expressing ARCPOMC neuron showing an increase in firing rate in response to CNO (10 μM). d, Expression of AAV-DIO-hM3Dq-mCherry (magenta) and Fos (green) in ARCPOMC neurons (right, overlay) in mice sacrificed 90 minutes after CNO administration. e, Effect of CNO/hM3Dq stimulation of ARCPOMC neurons on food intake. Mice from multiple litters, n = 5 animals. Paired two-tailed t-test: 24h: Saline (mean = 4.11, s.e.m. = 0.08843) versus CNO (mean = 3.354, s.e.m. = 0.2454): t(4) = 4.187, *P = 0.0138; 48h: Saline (mean = 7.89, s.e.m. = 0.2434) versus CNO (mean = 6.652, s.e.m. = 0.3109): t(4) = 6.420, **P = 0.0030. Data are presented as mean ± s.e.m. f, Schematic diagram of the Pomc-IRES-Cre allele. The IRES-Cre cassette was targeted just downstream of the stop codon in exon III of the Pomc gene. f, g, tdTomato expression in Pomc-IRES-Cre mice crossed with tdTomato reporter mice. h, tdTomato (magenta) and POMC expression (GFP) in the ARC (right, overlay). Yellow arrows indicate tdTomato-positive/POMC-negative neurons. i, Left, experimental schematic. Right, AAV-DIO-hM3Dq-mCherry expression in ARCPOMC neurons in Pomc-IRES-Cre mice as determined by mCherry immunolabeling. j, Expression of AAV-DIO-hM3Dq-mCherry (left) and Fos (middle) immunolabeling (right, overlay) in ARCPOMC neurons in Pomc-IRES-Cre mice sacrificed 90 minutes after CNO administration.

Supplementary Figure 2 Validation of hM3Dq/hM4Di expression and function in ARCVglut2 neurons.

a, Experimental schematic. b, c, Expression of AAV-DIO-hM3Dq-mCherry (b) or AAV-DIO-hM4Di-mCherry (c) in ARCVglut2 neurons in Vglut2-IRES-Cre mice as determined by mCherry immunolabeling. d, Representative trace of a cell-attached recording from a hM4Di-expressing ARCVglut2 neuron showing a decrease in firing rate in response to CNO (10 μM).

Supplementary Figure 3 Efferent targets of ARCVglut2 neurons.

a, Experimental schematic. AAV-DIO-synaptophysin-mCherry expresses the synaptic vesicle protein, synaptophysin fused with mCherry in a Cre-dependent fashion. b, Expression of AAV-DIO-synaptophysin-mCherry (ARCVglut2 neurons, magenta) and POMC (GFP) immunolabeling in the injection site (ARC). c, d, Expression of AAV-DIO-synaptophysin-mCherry and POMC immunolabeling in the ARC (c) and in the PVH (d). Yellow arrows indicate POMC-positive ARCVglut2 neurons (left) and POMC-positive terminals of ARCVglut2 neurons in the PVH (right). We found that a subset of ARCVglut2 neurons are POMC-positive (c). In contrast, we failed to detect POMC immunolabeling in the vast majority of ARCVglut2 terminals (d) in the PVH. Scale bars represent 20 μm. e, f, POMC (e) or AgRP (f) expression in ARCVglut2 projection sites as determined by expression of AAV-DIO-synaptophysin-mCherry and POMC (e) or AgRP (f) immunolabeling. ARCVglut2 neurons exhibit projections to the bed nucleus of the stria terminalis (BNST), preoptic area (POA), PVH, central nucleus of the amygdala, paraventricular thalamus (PVT), lateral hypothalamic area, dorsomedial hypothalamic nucleus, ventrolateral periaqueductal grey, lateral parabrachial nucleus and nucleus of solitary tractus. Qualitative assessment of fiber density demonstrated that the PVH, PVT, POA and BNST exhibited the most prominently labeled projections from ARCVglut2 neurons.

Supplementary Figure 4 Neuropeptide-defined subtypes of ARCVglut2 neurons and ARCVglut2 single-neuron gene expression of synapse-related genes.

a, Schematic of single-cell RNA-seq clustering analysis based on expression of 90 neuropeptide genes (see Supplemental Table 1). b, Two-dimensional representation of ARCVglut2 neurons (dots) by t-distributed stochastic neighbor embedding (tSNE). Subtypes are annotated according to neuropeptide gene markers. One subtype was not specifically marked by expression of a single neuropeptide (or other) gene and is annotated as “Other.” c, tSNE plot re-colored to show expression of prominent neuropeptide marker genes. Colors of dots indicate the level of expression relative to the maximal level for each gene. c, Expression of synapse-related genes in Pomc-positive (Pomc+) versus Pomc-negative (Pomc-) subsets of ARCVglut2 neurons. Sample size (cells): Pomc-positive/Pomc-negative, n = 10/13. Slc17a6: Pomc-positive (mean = 21.63, s.e.m. = 5.38) versus Pomc-negative (mean = 62.86, s.e.m. = 11.2): Unpaired two-tailed t-test; t(21) = 3.016, **P = 0.0066. Syt1: Pomc-positive (mean = 0.37, s.e.m. = 0.209) versus Pomc-negative (mean = 1.23, s.e.m. = 0.2662): Two-tailed Mann-Whitney test; U = 21, **P = 0.0052. Cplx1: Pomc-positive (mean = 3.412, s.e.m. = 2.054) versus Pomc-negative (mean = 20.26, s.e.m. = 5.574): Two-tailed Mann-Whitney test; U = 20, **P = 0.0041. Data are presented as mean ± s.e.m.

Supplementary Figure 5 ARCKiss1 neurons synaptically engage a small number of PVH neurons and do not influence food intake.

a, Cre-dependent ChR2 fused to a fluorescent protein (AAV-DIO-ChR2-mCherry) was unilaterally targeted to ARCKiss1 neurons. Images show injection site and terminal field in the PVH of ARCKiss1 neurons. Right, schematic shows connection being tested. Representative traces showing assessment of light-evoked EPSCs recorded from an unidentified PVH (uPVH) neuron. b, Expression of AAV-DIO-hM3Dq-mCherry in ARCKiss1 neurons in Kiss1-Cre mice as determined by mCherry immunolabeling. c, Expression of AAV-DIO-hM3Dq-mCherry (left) and Fos (middle) immunolabeling in ARCKiss1 neurons (right, overlay) in mice sacrificed 90 minutes after CNO administration. d, Effect of CNO/hM3Dq stimulation of ARCKiss1 neurons on dark-cycle food intake. Mice from multiple litters, n = 5 animals. Repeated measures two-way ANOVA: Treatment F(1,4) = 0.8651, P = 0.4050; time F(4,16) = 147.8, P < 0.0001; interaction F(4, 12) = 0.4285, P = 0.7860. Data are presented as mean ± s.e.m.

Supplementary Figure 6 CRACM mapping and validation of hM3Dq/ChR2 expression in ARCOxtr neurons.

a, Top, schematic shows connections being tested. Bottom, representative traces showing assessment of light-evoked IPSCs recorded from an uPVH neuron. No PVH-projecting ARCOxtr neurons are GABAergic. b, Expression of AAV-DIO-hM3Dq-mCherry in ARCOxtr neurons as determined by mCherry immunolabeling. c, Expression of AAV-DIO-hM3Dq-mCherry (left) and Fos (middle) immunolabeling in ARCOxtr neurons (right, overlay) in mice sacrificed 90 minutes after CNO administration. d, CNO/hM3Dq stimulation of ARCOxtr reduces refeeding following 24 hours fasting. Mice from multiple litters, n = 5 animals. Repeated measures two-way ANOVA: Treatment F(1,4) = 10.91, P = 0.0298; time F(4,16) = 64.22, P < 0.0001; interaction F(4, 16) = 6.976, P = 0.0019. Sidaks multiple comparisons test: 1h, P = 0.2882; 2h, **P = 0.0018; 3h, ****P = 0.0003; 4h, ****P < 0.0001. Data are presented as mean ± s.e.m. e, Expression of AAV-DIO-ChR2-mCherry (left) in ARCOxtr neurons and placement of optical fiber above the PVH (right).

Supplementary Figure 7 CRACM mapping to evaluate ARC →; PVH connectivity.

a, Left, Cre-independent ChR2 fused to a fluorescent protein (AAV-ChR2-mCherry) was unilaterally targeted to ARC neurons. Images show expression of AAV-ChR2-mCherry in the injection site and terminal field in the PVH. PVHMC4R neurons were transduced with AAV-DIO-GFP. Scale bars represent 100 μm. Right, schematic shows connections being tested (top) and representative traces of light-evoked EPSCs recorded from a PVHMC4R neuron before (black) and after (grey) bath application of the AMPAR antagonist CNQX. ARC, ARCGlutamatergic. b, c, Top, schematics show connections being tested. Bottom, representative traces showing assessment of light-evoked EPSCs recorded from PVHMC4R-negative (MC4R-) neurons.

Supplementary Figure 8 α-MSH effects on electrically evoked EPSCs and schematic diagram of the PomcloxP allele.

a, Left, Representative traces of electrically-evoked AMPAR-mediated EPSCs (black) and NMDAR-mediated EPSCs (grey) recorded under control conditions (top) and with α-MSH treatment (bottom). Right, Bar graph shows the effect of α-MSH on electrically evoked AMPAR/NMDAR ratios. Sample size (cells): Control/α-MSH, n = 5/5. Unpaired two-tailed t-test: Control (mean = 1.077, s.e.m. = 0.2054) versus α-MSH (mean = 2.465, s.e.m. = 0.548): t(8) = 2.373, *P = 0.0451. b, Representative traces of electrically-evoked EPSCs (left) and summary of the effects (right) of α-MSH on paired pulse ratio (PPR). Sample size (cells): Control/α-MSH, n = 9/10. Unpaired two-tailed t-test: Control (mean = 0.9177, s.e.m. = 0.1233) versus α-MSH (mean = 0.9629, s.e.m. = 0.06044): t(17) = 0.3402, P = 0.7379. Recordings were performed from PVHMC4R neurons identified in Mc4r-t2a-Cre::tdTomato mice. 10 slices from 5 mice; mice from multiple litters. Data are presented as mean ± s.e.m. c, Schematic shows the Pomclox allele. Mice with lox sites flanking exon III of the Pomc gene were generated using gene targeting in ES cells.

Supplementary Figure 9 Schematic summarizing the main findings.

Current models for homeostatic control of hunger / satiety lack a rapidly-acting satiety mechanism that is temporally analogous to that provided by rapidly-acting, hunger-promoting ARCAgRP neurons. Our findings report the discovery of this missing component, namely glutamate-releasing oxytocin receptor-expressing neurons in the arcuate nucleus. These neurons converge with ARCAgRP and ARCPOMC neurons on downstream targets and rapidly increase / decrease satiety when experimentally activated / inhibited. Thus, in contrast to the hunger-promoting system where slow- (AgRP) and rapid-acting signals (GABA and NPY) are conveyed by one group of neurons (ARCAgRP neurons), the counteracting satiety-promoting system is split into two parallel-projecting neurons, the previously known ARCPOMC neurons which deliver slow-acting α-MSH, and the previously unknown ARCOxtr neurons which release the rapid-acting transmitter, glutamate. Importantly, these two satiety neurons interact as α-MSH released by ARCPOMC neurons postsynaptically increases glutamatergic transmission across the ARCOxtr→PVHMC4R synapse. This missing rapid-acting component, with ARCPOMC neurons, now provides the full “yang” to the ARCAgRP neuron’s “yin”. Our work also provides a mechanism for α-MSH /MC4R regulation of satiety – plasticity of the novel ARCOxtr→PVHMC4R synapse.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Table 1 (PDF 2001 kb)

Supplementary Methods Checklist (PDF 549 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fenselau, H., Campbell, J., Verstegen, A. et al. A rapidly acting glutamatergic ARC→PVH satiety circuit postsynaptically regulated by α-MSH. Nat Neurosci 20, 42–51 (2017). https://doi.org/10.1038/nn.4442

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4442

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