DOI QR코드

DOI QR Code

Network of hypothalamic neurons that control appetite

  • Sohn, Jong-Woo (Department of Biological Sciences, Korea Advanced Institute of Science and Technology)
  • Received : 2014.10.06
  • Published : 2015.04.30

Abstract

The central nervous system (CNS) controls food intake and energy expenditure via tight coordinations between multiple neuronal populations. Specifically, two distinct neuronal populations exist in the arcuate nucleus of hypothalamus (ARH): the anorexigenic (appetite-suppressing) pro-opiomelanocortin (POMC) neurons and the orexigenic (appetite-increasing) neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons. The coordinated regulation of neuronal circuit involving these neurons is essential in properly maintaining energy balance, and any disturbance therein may result in hyperphagia/obesity or hypophagia/starvation. Thus, adequate knowledge of the POMC and NPY/AgRP neuron physiology is mandatory to understand the pathophysiology of obesity and related metabolic diseases. This review will discuss the history and recent updates on the POMC and NPY/AgRP neuronal circuits, as well as the general anorexigenic and orexigenic circuits in the CNS. [BMB Reports 2015; 48(4): 229-233]

Keywords

INTRODUCTION

Hypothalamus is a key brain area that regulates homeostasis. In particular, specific areas of hypothalamus are believed to control feeding behavior. The classic experiments by Hetherington and Ranson established the ventromedial hypothalamus as the appetite-suppressing (or anorexigenic) center (1), and later experiments demonstrated that lateral hypothalamic area (LHA) is the appetite-increasing (or orexigenic) center (2). These results suggested that a specific hypothalamic area may regulate feeding, and subsequent studies attempted to revise this concept by using more refined neuroanatomical methods. In recent years, the development of mouse genetics and other techniques such as electrophysiology, optogenetics, and chemogenetics has led us to gain more detailed information on the identity of specific neurons that affect feeding behavior. Specifically, a large body of information is currently available on the appetite-regulating role of two distinct neuronal populations within the ventral medial part of hypothalamus.

 

THE ARCUATE NUCLEUS OF HYPOTHALAMUS

The arcuate nucleus of hypothalamus (ARH) is undoubtedly one of the best-characterized brain regions as it is related to the control of feeding behavior. This is in large part due to the presence of two distinct neuronal populations, which have opposite effects on the feeding behavior: the anorexigenic pro-opiomelanocortin (POMC) neurons and the orexigenic neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons. These neurons are well-positioned to receive information from peripheral organs; the ARH resides in the ventral medial part of hypothalamus, which receives rich blood supply due to its proximity to the median eminence. Thus, information from peripheral organs may easily access the POMC and NPY/AgRP neurons. In addition, they receive intensive input from multiple parts of the central nervous system (CNS). Therefore, POMC and NPY/AgRP neurons are in a good position to integrate peripheral and central inputs to produce a central command for feeding behavior. Indeed, the activity of POMC and NPY/AgRP neurons is modulated by multiple neurotransmitters and/or hormones. For instance, the anorexigenic effects of serotonin and the adipocyte-derived hormone leptin are considered to be at least in part mediated by excitation of POMC neurons and suppression of NPY/AgRP neurons (3-6). Ghrelin, an orexigenic hormone released from gastric mucosa, was shown to suppress POMC neurons and excite NPY/AgRP neurons by indirect mechanisms (7-9). The effects of insulin on POMC neurons and NPY/AgRP neurons need to be clarified, as the experimental results are not consistent between studies from independent groups (6, 10, 11).

 

THE POMC NEURONS AND THE CENTRAL MELANOCORTIN PATHWAY

The anorexigenic effect of POMC neuron was evidenced by the hyperphagia and obesity observed in the POMC knockout mice (12). Recent studies employed optogenetic and chemogenetic stimulation methods to activate specific neuronal population, and demonstrated that direct activation of POMC neurons lead to suppression of food intake (13, 14). It is believed that POMC neurons suppress appetite by releasing α-melanocyte stimulating hormone (α-MSH) which is an agonist at the anorectic melanocortin-4 receptors (MC4Rs). Consistent with this idea, MC4R deficiency results in hyperphagia and obesity in mice (15-17). Importantly, human patients with mutations in Mc4r genes are also hyperphagic and obese (18, 19). Thus, the central melanocortin pathway that involves POMC neurons and the MC4R-expressing neurons represent a key anorexigenic circuit in the CNS.

One of the key anorexigenic signals that activate the central melanocortin pathway is serotonin (5-HT). Fenfluramine (d-Fen), which increases the availability of serotonin in brain, was an effective and widely-used prescription diet pill until it was withdrawn from the market due to serious cardiovascular side effects (20). The anorexigenic effect of serotonin is largely mediated by the serotonin 2C receptors (5-HT2CRs) expressed by the POMC neurons (21-23). Stimulation of 5-HT2CRs increases the activity of POMC neurons (21, 24, 25), which presumably increases α-MSH release. Consistently, the anorexigenic effects of d-Fen were demonstrated to be mediated by the MC4Rs expressed by the single-minded 1 (Sim1) neurons within the paraventricular nucleus of hypothalamus (PVH), as well as the 5-HT2CRs expressed by POMC neurons (26). Currently, d-Fen has been replaced by lorcaserin, a novel prescription diet pill and a specific agonist at 5-HT2CRs (27), which is expected to activate the central melanocortin pathway.

Unlike 5-HT2CRs, however, the deletion of leptin receptors (LepRs) specifically in POMC neurons does not increase food intake (28). In addition, the reactivation of LepRs specifically in POMC neurons of LepR-null mice does not rescue hyperphagia (29). Thus, while leptin activates POMC neurons, the anorexigenic effects of leptin are likely to be mediated by LepRs expressed in other parts of the brain, independent of the melanocortin system (30). In fact, leptin-induced activation of POMC neurons does not seem to activate central melanocortin pathways that suppress food intake. Instead, it seems that leptin excitation of POMC neurons activates central melanocortin pathways that increase energy expenditure (28, 29). Considering that energy expenditure is not affected by deletion or reactivation of 5-HT2CRs specifically in POMC neurons (22, 23), it is suspected that POMC neurons are heterogeneous in their response to leptin and serotonin. Consistent with this idea, the acute response of POMC neurons to leptin and mCPP (a 5-HT2CR agonist) is segregated to distinct subpopulations of POMC neurons (24). These results are intriguing since the metabolic effects of MC4Rs are mediated by different brain nuclei; MC4Rs expressed by the PVH decreases food intake, while MC4Rs expressed by the sympathetic neurons within the intermediolateral column (IML) of spinal cord increases energy expenditure (16, 31). Taken together, the “appetite-suppressing” POMC neurons may project to PVH, while the “energy-consuming” POMC neurons may project to the IML (Fig. 1).

Fig. 1.POMC neurons and the central melanocortin pathway. Two distinct populations of POMC neurons are present within the ARH; 5-HT2CR-expressing POMC neurons suppress food intake, and LepR-expressing POMC neurons increase energy expenditure. The effects of MC4Rs on energy balance are localized to different central nuclei; MC4Rs expressed by the PVH Sim1 neurons suppress food intake, and MC4Rs expressed by the IML sympathetic neurons increase energy expenditure. The solid black lines show possible connections between the specific POMC neurons and specific brain nuclei; the connections represented by the broken gray lines have not been ruled out. MC4R-expressing PVH glutamatergic neurons send axons to the neurons of the L-PBN. 3V: third ventricle, 5-HT2CR: serotonin 2C receptor, α-MSH: α-melanocyte stimulating hormone, ChAT: choline acetyltransferase, EE: energy expenditure, FI: food intake, IML: intermediolateral column of spinal cord, L-PBN: lateral parabrachial nucleus, LepR: leptin receotor, MC4R: melanocortin-4 receptor, ME: median eminence, POMC: pro-opiomelanocortin, PVH: paraventricular nucleus of hypothalamus, Sim1: single-minded 1.

 

THE PARAVENTRICULAR NUCLEUS OF HYPOTHALAMUS AND THE PARABRACHIAL NUCLEUS: ANOREXIGENIC CENTERS

The PVH and the parabrachial nucleus (PBN) of brainstem are representative anorexigenic centers in addition to POMC neurons and the central melanocortin pathways. Sim1 is a transcription factor that is critical for PVH development, and Sim 1 haploinsufficiency is associated with hyperphagia and obesity (32). Thus, a significant population of PVH neurons is considered to suppress food intake. Specifically, the oxytocin (OXT) neurons within the PVH are known to have loss-of-function mutations in Prader-Willi syndrome (33) and as a consequence of Sim1 deficiency (34, 35), both of which are characterized by hyperphagia. The MC4R-expressing glutamatergic neurons within the PVH, which may be considered as a part of the central melanocortin system, reduce food intake (36). Intriguingly, the MC4R-expressing PVH neurons were found to be distinct subpopulation from the OXT, corticotropin-releasing hormone (CRH), arginine vasopressin (AVP), and prodynorphin (Pdyn) neurons within the PVH (36). In addition, Sim1 neurons that express either thyrotropin-releasing hormone (TRH) or pituitary adenylate cyclase-activating peptide (PACAP) were found to send excitatory input to the orexigenic NPY/AgRP neurons within the ARH (37). Thus, some PVH neurons may not be anorexigenic as originally suggested, but they may have “orexigenic” effects.

The PBN is a pontine nucleus located adjacent to the superior cerebellar peduncle that contains subpopulation of neurons suppressing appetite (38-40). The PBN neurons receive gluatamatergic “satiety” signals from the nucleus tractus solitarius (NTS) neurons located in the medulla oblongata (38), which is considered to be the neural correlates of postprandial satiety. Consistent with the anorexigenic role of PBN, the MC4R-expressing PVH glutamatergic neurons send axons to the lateral PBN (36) (Fig. 1). Recently, it was demonstrated that the calcitonin gene-related peptide (CGRP) neurons within the external lateral subdivision of PBN project to the central amygdala to suppress food intake (41). Another recent study also demonstrated that neurons expressing protein kinase C delta (PKCδ) mediate the anorexigenic effects (42).

 

THE NPY/AGRP NEURONS AND THE OREXIGENIC PATHWAY

The NPY/AgRP neurons within the ARH are probably the most established orexigenic population in the CNS. However, earlier studies reported that mice with NPY and/or AgRP deficiency have normal food intake and body weight (43). Similarly, ablation of AgRP neurons during development resulted in normal body weight (44, 45). More recent studies used toxins to ablate the NPY/AgRP neurons in adults and confirmed the orexigenic role of these neurons (38, 39, 46). Acute activation of NPY/AgRP neurons using optogenetic or pharmacogenetic stimulation methods also resulted in robust increase in food intake (13, 47). These series of experiments have established the NPY/AgRP neurons as the major orexigenic population in the CNS.

The NPY/AgRP neurons release the neuropeptide NPY (agonist at the Y receptors), AgRP (inverse agonist at the MC4Rs), and the inhibitory neurotransmitter GABA. The metabolic effects of NPY on Y receptors are somewhat complicated, and many are not consistent with the orexigenic role of NPY/AgRP neurons; this issue was discussed in more detail in a previous review article (48). The ability of AgRP to antagonize the central melanocortin pathways received much attention, since this was considered to be the underlying mechanism of orexigenic effects. However, the orexigenic effects of NPY/AgRP neurons are not affected by the deletion of MC4Rs (47), and it is now regarded that the NPY/AgRP neurons increase food intake independent of the central melanocortin system. Currently, GABA release is believed to mediate most of the orexigenic effects of the NPY/AgRP neurons. For instance, deletion of vesicular GABA transporter (vgat) genes specifically in the AgRP neurons leads to lean phenotype (49), which confirms the importance of GABAergic neurotransmission in the orexigenic effects.

Multiple “anorexigenic” neuronal populations within CNS receive the “inhibitory” GABAergic input from the NPY/AgRP neurons (Fig. 2). The ARH POMC neurons receive GABAergic input from the NPY/AgRP neurons, which form a local circuit of appetite regulation within the ARH (3). Indeed, when the vgat gene is deleted in the NPY/AgRP neurons, the frequency of inhibitory postsynaptic current (IPSC) is not increased by ghrelin which excites the NPY/AgRP neurons (49). The PVH OXT neurons and Sim1 neurons (which do not express either PACAP or TRH) also receive GABAergic input from the NPY/AgRP neurons (37, 50). Finally, the neurons within the PBN receive GABAergic input from the NPY/AgRP neurons (38, 39). Thus, it is suggested that the orexigenic pathways involving the GABAergic neurotramsission from the NPY/AgRP neurons increase food intake by the inhibition of the anorexigenic centers in the CNS.

Fig. 2.NPY/AgRP neurons and the orexigenic pathway. NPY/AgRP neurons have axon terminals that release GABA (an inhibitory neurotransmitter) to suppress the POMC neurons, PVH OXT neurons, and neurons within the L-PBN. PVH neurons that express PACAP/TRH have axon terminals that release glutamate (an excitatory neurotransmitter) to activate NPY/AgRP neurons. Functional connections between NPY/AgRP neurons and PVH neurons that express CRH/AVP have not been established (dotted lines). AgRP: agouti-related peptide, AVP: arginine vasopressin, CRH: corticotropin-releasing hormone, L-PBN: lateral parabrachial nucleus, NPY: neuropeptide Y, OXT: oxytocin, PACAP: pituitary adenylate cyclase-activating peptide, POMC: pro-opiomelanocortin, PVH: paraventricular nucleus of hypothalamus, TRH: thyrotropin-releasing hormone.

 

CONCLUDING REMARKS AND FUTURE DIRECTIONS

Recent development of genetic technologies (e.g. mouse genetics, optogenetics, and chemogenetics) was combined with classical methodologies (e.g. neuroanatomy and electrophysiology) to allow us to have a refined understanding of neuronal circuits that regulate feeding behavior. As a result, we now understand that feeding is regulated by “specific neurons within specific nucleus” rather than “centers” in brain. It is important to note that PVH, an anorexigenic center, was recently suggested to contain potentially orexigenic neurons (PACAP and TRH neurons) which send excitatory input to NPY/AgRP neurons (37). We still do not completely understand whether other specific PVH neurons (e.g. CRH, AVP, and Pdyn neurons) have orexigenic effects. In addition, the role of specific neuronal populations within PBN is just beginning to be identified (41). Thus, it should be important to study the function of genetically-identified neurons, both in vitro and in vivo, to have a more refined understanding on the neuronal circuits that regulate feeding behavior.

References

  1. Hetherington AW and Ranson SW (1940) Hypothalamic lesions and adiposity in the rat. Anat Rec 78, 149-172 https://doi.org/10.1002/ar.1090780203
  2. Anand BK and Brobeck JR (1951) Localization of a "feeding center" in the hypothalamus of the rat. Proc Soc Exp Biol Med 77, 323-324 https://doi.org/10.3181/00379727-77-18766
  3. Cowley MA, Smart JL, Rubinstein M et al (2001) Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411, 480-484 https://doi.org/10.1038/35078085
  4. Hill JW, Williams KW, Ye C et al (2008) Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice. J Clin Invest 118, 1796-1805 https://doi.org/10.1172/JCI32964
  5. van den Top M, Lee K, Whyment AD, Blanks AM and Spanswick D (2004) Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate nucleus. Nat Neurosci 7, 493-494 https://doi.org/10.1038/nn1226
  6. Williams KW, Margatho LO, Lee CE et al (2010) Segregation of acute leptin and insulin effects in distinct populations of arcuate proopiomelanocortin neurons. J Neurosci 30, 2472-2479 https://doi.org/10.1523/JNEUROSCI.3118-09.2010
  7. Cowley MA, Smith RG, Diano S et al (2003) The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37, 649-661 https://doi.org/10.1016/S0896-6273(03)00063-1
  8. van den Pol AN, Yao Y, Fu LY et al (2009) 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 https://doi.org/10.1523/JNEUROSCI.3249-08.2009
  9. Yang Y, Atasoy D, Su HH and Sternson SM (2011) Hunger states switch a flip-flop memory circuit via a synaptic AMPK-dependent positive feedback loop. Cell 146, 992-1003 https://doi.org/10.1016/j.cell.2011.07.039
  10. Qiu J, Zhang C, Borgquist A et al (2014) Insulin excites anorexigenic proopiomelanocortin neurons via activation of canonical transient receptor potential channels. Cell Metab 19, 682-693 https://doi.org/10.1016/j.cmet.2014.03.004
  11. Al-Qassab H, Smith MA, Irvine EE et al (2009) Dominant role of the p110beta isoform of PI3K over p110alpha in energy homeostasis regulation by POMC and AgRP neurons. Cell Metab 10, 343-354 https://doi.org/10.1016/j.cmet.2009.09.008
  12. Yaswen L, Diehl N, Brennan MB and Hochgeschwender U (1999) Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nat Med 5, 1066-1070 https://doi.org/10.1038/12506
  13. Aponte Y, Atasoy D and Sternson SM (2011) AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat Neurosci 14, 351-355 https://doi.org/10.1038/nn.2739
  14. Zhan C, Zhou J, Feng Q et al (2013) Acute and Long-Term Suppression of Feeding Behavior by POMC Neurons in the Brainstem and Hypothalamus, Respectively. J Neurosci 33, 3624-3632 https://doi.org/10.1523/JNEUROSCI.2742-12.2013
  15. Huszar D, Lynch CA, Fairchild-Huntress V et al (1997) Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88, 131-141 https://doi.org/10.1016/S0092-8674(00)81865-6
  16. Balthasar N, Dalgaard LT, Lee CE et al (2005) Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 123, 493-505 https://doi.org/10.1016/j.cell.2005.08.035
  17. Fan W, Boston BA, Kesterson RA, Hruby VJ and Cone RD (1997) Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385, 165-168 https://doi.org/10.1038/385165a0
  18. Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG and O'Rahilly S (1998) A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 20, 111-112 https://doi.org/10.1038/2404
  19. Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T and O'Rahilly S (2003) Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 348, 1085-1095 https://doi.org/10.1056/NEJMoa022050
  20. Connolly HM, Crary JL, McGoon MD et al (1997) Valvular heart disease associated with fenfluramine-phentermine. New Engl J Med 337, 581-588 https://doi.org/10.1056/NEJM199708283370901
  21. Heisler LK, Cowley MA, Tecott LH et al (2002) Activation of central melanocortin pathways by fenfluramine. Science 297, 609-611 https://doi.org/10.1126/science.1072327
  22. Xu Y, Jones JE, Kohno D et al (2008) 5-HT2CRs expressed by pro-opiomelanocortin neurons regulate energy homeostasis. Neuron 60, 582-589 https://doi.org/10.1016/j.neuron.2008.09.033
  23. Berglund ED, Liu C, Sohn JW et al (2013) Serotonin 2C receptors in pro-opiomelanocortin neurons regulate energy and glucose homeostasis. J Clin Invest 123, 5061-5070 https://doi.org/10.1172/JCI70338
  24. Sohn JW, Xu Y, Jones JE, Wickman K, Williams KW and Elmquist JK (2011) Serotonin 2C Receptor Activates a Distinct Population of Arcuate Pro-opiomelanocortin Neurons via TRPC Channels. Neuron 71, 488-497 https://doi.org/10.1016/j.neuron.2011.06.012
  25. Roepke TA, Smith AW, Ronnekleiv OK and Kelly MJ (2012) Serotonin 5-HT2C receptor-mediated inhibition of the M-current in hypothalamic POMC neurons. Am J Physiol Endocrinol Metab 302, E1399-1406 https://doi.org/10.1152/ajpendo.00565.2011
  26. Xu Y, Jones JE, Lauzon DA et al (2010) A serotonin and melanocortin circuit mediates D-fenfluramine anorexia. J Neurosci 30, 14630-14634 https://doi.org/10.1523/JNEUROSCI.5412-09.2010
  27. Smith SR, Weissman NJ, Anderson CM et al (2010) Multicenter, placebo-controlled trial of lorcaserin for weight management. New Engl J Med 363, 245-256 https://doi.org/10.1056/NEJMoa0909809
  28. Balthasar N, Coppari R, McMinn J et al (2004) Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 42, 983-991 https://doi.org/10.1016/j.neuron.2004.06.004
  29. Berglund ED, Vianna CR, Donato J Jr et al (2012) Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J Clin Invest 122, 1000-1009 https://doi.org/10.1172/JCI59816
  30. Scott MM, Williams KW, Rossi J, Lee CE and Elmquist JK (2011) Leptin receptor expression in hindbrain Glp-1 neurons regulates food intake and energy balance in mice. J Clin Invest 121, 2413-2421 https://doi.org/10.1172/JCI43703
  31. Rossi J, Balthasar N, Olson D et al (2011) Melanocortin-4 receptors expressed by cholinergic neurons regulate energy balance and glucose homeostasis. Cell Metab 13, 195-204 https://doi.org/10.1016/j.cmet.2011.01.010
  32. Michaud JL, Boucher F, Melnyk A et al (2001) Sim1 haploinsufficiency causes hyperphagia, obesity and reduction of the paraventricular nucleus of the hypothalamus. Hum Mol Genet 10, 1465-1473 https://doi.org/10.1093/hmg/10.14.1465
  33. Swaab DF, Purba JS and Hofman MA (1995) Alterations in the hypothalamic paraventricular nucleus and its oxytocin neurons (putative satiety cells) in Prader-Willi syndrome: a study of five cases. J Clini Endocrin Metab 80, 573-579
  34. Kublaoui BM, Gemelli T, Tolson KP, Wang Y, and Zinn AR (2008) Oxytocin deficiency mediates hyperphagic obesity of Sim1 haploinsufficient mice. Mol Endocrinol 22, 1723-1734 https://doi.org/10.1210/me.2008-0067
  35. Holder JL Jr, Butte NF and Zinn AR (2000) Profound obesity associated with a balanced translocation that disrupts the SIM1 gene. Hum Mol Genet 9, 101-108 https://doi.org/10.1093/hmg/9.1.101
  36. Shah BP, Vong L, Olson DP et al (2014) MC4R-expressing glutamatergic neurons in the paraventricular hypothalamus regulate feeding and are synaptically connected to the parabrachial nucleus. Proc Natl Acad Sci U S A 111, 13193-13198 https://doi.org/10.1073/pnas.1407843111
  37. Krashes MJ, Shah BP, Madara JC et al (2014) An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature 507, 238-242 https://doi.org/10.1038/nature12956
  38. Wu Q, Clark MS and Palmiter RD (2012) Deciphering a neuronal circuit that mediates appetite. Nature 483, 594-597 https://doi.org/10.1038/nature10899
  39. Wu Q, Boyle MP and Palmiter RD (2009) Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell 137, 1225-1234 https://doi.org/10.1016/j.cell.2009.04.022
  40. Becskei C, Grabler V, Edwards GL, Riediger T and Lutz TA (2007) Lesion of the lateral parabrachial nucleus attenuates the anorectic effect of peripheral amylin and CCK. Brain Res 1162, 76-84 https://doi.org/10.1016/j.brainres.2007.06.016
  41. Carter ME, Soden ME, Zweifel LS and Palmiter RD (2013) Genetic identification of a neural circuit that suppresses appetite. Nature 503, 111-114 https://doi.org/10.1038/nature12596
  42. Cai H, Haubensak W, Anthony TE, Anderson DJ (2014) Central amygdala PKC-delta(+) neurons mediate the influence of multiple anorexigenic signals. Nat Neurosci 17, 1240-1248 https://doi.org/10.1038/nn.3767
  43. Qian S, Chen H, Weingarth D et al (2002) Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Mol Cell Biol 22, 5027-5035 https://doi.org/10.1128/MCB.22.14.5027-5035.2002
  44. Luquet S, Perez FA, Hnasko TS and Palmiter RD (2005) NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310, 683-685 https://doi.org/10.1126/science.1115524
  45. Luquet S, Phillips CT and Palmiter RD (2007) NPY/AgRP neurons are not essential for feeding responses to glucoprivation. Peptides 28, 214-225 https://doi.org/10.1016/j.peptides.2006.08.036
  46. Gropp E, Shanabrough M, Borok E et al (2005) Agouti-related peptide-expressing neurons are mandatory for feeding. Nat Neurosci 8, 1289-1291 https://doi.org/10.1038/nn1548
  47. Krashes MJ, Koda S, Ye C et al (2011) Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest 121, 1424-1428 https://doi.org/10.1172/JCI46229
  48. Sohn JW, Elmquist JK and Williams KW (2013) Neuronal circuits that regulate feeding behavior and metabolism. Trends Neurosci 36, 504-512 https://doi.org/10.1016/j.tins.2013.05.003
  49. Tong Q, Ye CP, Jones JE, Elmquist JK and Lowell BB (2008) Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat Neurosci 11, 998-1000 https://doi.org/10.1038/nn.2167
  50. Atasoy D, Betley JN, Su HH and Sternson SM (2012) Deconstruction of a neural circuit for hunger. Nature 488, 172-177 https://doi.org/10.1038/nature11270

Cited by

  1. A brief review of salient factors influencing adult eating behaviour 2017, https://doi.org/10.1017/S0954422417000099
  2. Endocrine disruptors and obesity vol.11, pp.11, 2015, https://doi.org/10.1038/nrendo.2015.163
  3. Circadian Disruptions in the Myshkin Mouse Model of Mania Are Independent of Deficits in Suprachiasmatic Molecular Clock Function 2017, https://doi.org/10.1016/j.biopsych.2017.04.018
  4. Evidence for the involvement of neuropeptide Y in the antidepressant effect of imipramine in type 2 diabetes vol.1646, 2016, https://doi.org/10.1016/j.brainres.2016.05.035
  5. Biotin augments acetyl CoA carboxylase 2 gene expression in the hypothalamus, leading to the suppression of food intake in mice vol.476, pp.3, 2016, https://doi.org/10.1016/j.bbrc.2016.04.152
  6. The central effect of 3-iodothyronamine on brain neuropeptides in mice vol.26, pp.5, 2017, https://doi.org/10.1007/s00580-017-2495-2
  7. Ambient temperature modulates the effects of the Prader-Willi syndrome candidate gene Snord116 on energy homeostasis vol.61, 2017, https://doi.org/10.1016/j.npep.2016.10.006
  8. Neuropeptide FF increases M2 activation and self-renewal of adipose tissue macrophages vol.127, pp.7, 2017, https://doi.org/10.1172/JCI90152
  9. Cyclocarya paliurus (Batal.) Ijinskaja Aqueous Extract (CPAE) Ameliorates Obesity by Improving Insulin Signaling in the Hypothalamus of a Metabolic Syndrome Rat Model vol.2017, 2017, https://doi.org/10.1155/2017/4602153
  10. Identification of a small-molecule ligand that activates the neuropeptide receptor GPR171 and increases food intake vol.9, pp.430, 2016, https://doi.org/10.1126/scisignal.aac8035
  11. Obesity: Current and potential pharmacotherapeutics and targets vol.170, 2017, https://doi.org/10.1016/j.pharmthera.2016.10.015
  12. Intranasal gene delivery for treating Parkinson’s disease: overcoming the blood–brain barrier vol.12, pp.12, 2015, https://doi.org/10.1517/17425247.2015.1069815
  13. Metabolism disrupting chemicals and metabolic disorders vol.68, 2017, https://doi.org/10.1016/j.reprotox.2016.10.001
  14. Hunger and thirst interact to regulate ingestive behavior in flies and mammals vol.39, pp.5, 2017, https://doi.org/10.1002/bies.201600261
  15. The Neuroendocrine Regulation of Food Intake in Fish: A Review of Current Knowledge vol.10, 2016, https://doi.org/10.3389/fnins.2016.00540
  16. Acute and Long-Term Impact of High-Protein Diets on Endocrine and Metabolic Function, Body Composition, and Exercise-Induced Adaptations vol.36, pp.4, 2017, https://doi.org/10.1080/07315724.2016.1274691
  17. Neuroinflammatory and autonomic mechanisms in diabetes and hypertension vol.311, pp.1, 2016, https://doi.org/10.1152/ajpendo.00012.2016
  18. Stimulation of the hypothalamic arcuate nucleus increases brown adipose tissue nerve activity via hypothalamic paraventricular and dorsomedial nuclei vol.311, pp.2, 2016, https://doi.org/10.1152/ajpheart.00176.2016
  19. 4-hydroxy-3-methoxycinnamic acid regulates orexigenic peptides and hepatic glucose homeostasis through phosphorylation of FoxO1 vol.50, pp.2, 2018, https://doi.org/10.1038/emm.2017.253
  20. Maternal Diabetes and Fetal Programming Toward Neurological Diseases: Beyond Neural Tube Defects vol.9, pp.1664-2392, 2018, https://doi.org/10.3389/fendo.2018.00664
  21. Brain SIRT1 Mediates Metabolic Homeostasis and Neuroprotection vol.9, pp.1664-2392, 2018, https://doi.org/10.3389/fendo.2018.00702
  22. Making way for suppressing the FGF19/FGFR4 axis in cancer vol.10, pp.20, 2018, https://doi.org/10.4155/fmc-2018-0099
  23. An overview of energy and metabolic regulation pp.1869-1889, 2018, https://doi.org/10.1007/s11427-018-9371-4
  24. Central regulation of food intake in fish: an evolutionary perspective vol.60, pp.4, 2018, https://doi.org/10.1530/JME-17-0320
  25. Morphofunctional Organization of the Connections From the Medial and Intermediate Parts of the Central Nucleus of the Amygdala Into Distinct Divisions of the Lateral Hypothalamic Area in the Rat vol.9, pp.1664-2295, 2018, https://doi.org/10.3389/fneur.2018.00688
  26. Chronic treatment with tributyltin induces sexually dimorphic alterations in the hypothalamic POMC system of adult mice pp.1432-0878, 2018, https://doi.org/10.1007/s00441-018-2896-9
  27. Integrating Thyroid Hormone Signaling in Hypothalamic Control of Metabolism: Crosstalk Between Nuclear Receptors vol.19, pp.7, 2018, https://doi.org/10.3390/ijms19072017
  28. Voglibose-mediated alterations in neurometabolomic profiles in the hypothalamus of high-fat diet-fed mice pp.1476-8305, 2018, https://doi.org/10.1080/1028415X.2018.1443995
  29. Obesity and cahexia as the first manifestations of craniopharingioma vol.15, pp.1, 2018, https://doi.org/10.14341/omet2018143-49
  30. The Leptin, Dopamine and Serotonin Receptors in Hypothalamic POMC-Neurons of Normal and Obese Rodents vol.43, pp.4, 2018, https://doi.org/10.1007/s11064-018-2485-z
  31. Intestinal epithelial N-acylphosphatidylethanolamine phospholipase D links dietary fat to metabolic adaptations in obesity and steatosis vol.10, pp.1, 2019, https://doi.org/10.1038/s41467-018-08051-7