3,603
Views
100
CrossRef citations to date
0
Altmetric
Comprehensive Review

The role of leptin in health and disease

&
Pages 258-291 | Received 21 Dec 2016, Accepted 02 May 2017, Published online: 01 Aug 2017

References

  • Margetic S, Gazzola C, Pegg GG, Hill RA. Leptin: a review of its peripheral actions and interactions. Int J Obes Relat Metab Disord. 2002;26:1407-1433; PMID:12439643. doi:10.1038/sj.ijo.0802142.
  • Sinha MK, Ohannesian JP, Heiman ML, Kriauciunas A, Stephens TW, Magosin S, Marco C, Caro JF. Nocturnal rise of leptin in lean, obese, and non-insulin-dependent diabetes mellitus subjects. J Clin Invest. 1996;97:1344-1347; PMID:8636448. doi:10.1172/JCI118551.
  • Saad MF, Damani S, Gingerich RL, Riad-Gabriel MG, Khan A, Boyadjian R, Jinagouda SD, el-Tawil K, Rude RK, Kamdar V. Sexual dimorphism in plasma leptin concentration. J Clin Endocrinol Metab. 1997;82:579-584; PMID:9024258..
  • Tsai M, Asakawa A, Amitani H, Inui A. Stimulation of leptin secretion by insulin. Indian J Endocrinol Metab. 2012;16:S543-S548; PMID:23565488. doi:10.4103/2230-8210.105570.
  • Vital P, Larrieta E, Hiriart M. Sexual dimorphism in insulin sensitivity and susceptibility to develop diabetes in rats. J Endocrinol. 2006;190:425-432; PMID:16899575. doi:10.1677/joe.1.06596.
  • Leal-Cerro A, Soto A, Martinez MA, Dieguez C, Casanueva FF. Influence of cortisol status on leptin secretion. Pituitary. 2001;4:111-116; PMID:11824503. doi:10.1023/A:1012903330944.
  • Coleman DL. Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia. 1978;14:141-148; PMID:350680. doi:10.1007/BF00429772.
  • Coleman DL. A historical perspective on leptin. Nat Med. 2010;16:1097-1099; PMID:20930752. doi:10.1038/nm1010-1097.
  • Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science. 1966;153:1127-1128; PMID:5918576. doi:10.1126/science.153.3740.1127.
  • Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house mouse. J Hered. 1950;41:317-318; PMID:14824537. doi:10.1093/oxfordjournals.jhered.a106073.
  • Coleman DL. Effects of parabiosis of obese with diabetes and normal mice. Diabetologia. 1973;9:294-298; PMID:4767369. doi:10.1007/BF01221857.
  • Coleman DL, Hummel KP. Effects of parabiosis of normal with genetically diabetic mice. Am J Physiol. 1969;217:1298-1304; PMID:5346292.
  • Coleman DL, Hummel KP. The influence of genetic background on the expression of the obese (Ob) gene in the mouse. Diabetologia. 1973;9:287-293; PMID:4588246. doi:10.1007/BF01221856.
  • Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425-432; PMID:7984236. doi:10.1038/372425a0.
  • Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, et al. Identification and expression cloning of a leptin receptor, OB-R. Cell. 1995;83:1263-1271; PMID:8548812. doi:10.1016/0092-8674(95)90151-5.
  • Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, et al. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell. 1996;84:491-495; PMID:8608603. doi:10.1016/S0092-8674(00)81294-5.
  • Zhang J, Scarpace PJ. The soluble leptin receptor neutralizes leptin-mediated STAT3 signalling and anorexic responses in vivo. Br J Pharmacol. 2009;158:475-482; PMID:19422379. doi:10.1111/j.1476-5381.2009.00246.x.
  • Hileman SM, Pierroz DD, Masuzaki H, Bjorbak C, El-Haschimi K, Banks WA, Flier JS. Characterizaton of short isoforms of the leptin receptor in rat cerebral microvessels and of brain uptake of leptin in mouse models of obesity. Endocrinology. 2002;143:775-783; PMID:11861497. doi:10.1210/endo.143.3.8669.
  • Bjorbaek C, Uotani S, da Silva B, Flier JS. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem. 1997;272:32686-32695; PMID:9405487. doi:10.1074/jbc.272.51.32686.
  • Chua SC, Koutras IK, Han L, Liu S-M, Kay J, Young SJ, Chung WK, Leibel RL. Fine structure of the murine leptin receptor gene: splice site suppression is required to form two alternatively spliced transcripts. Genomics. 1997;45:264-270; PMID:9344648. doi:10.1006/geno.1997.4962.
  • Tartaglia LA. The leptin receptor. J Biol Chem. 1997;272:6093-6096; PMID:9102398. doi:10.1074/jbc.272.10.6093.
  • Vaisse C, Halaas JL, Horvath CM, Darnell JE Jr, Stoffel M, Friedman JM. Leptin activation of Stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice. Nat Genet. 1996;14:95-97; PMID:8782827. doi:10.1038/ng0996-95.
  • Chua SC, Chung WK, Wu-Peng XS, Zhang Y, Liu S-M, Tartaglia L, Leibel RL. Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (Leptin) receptor. Science. 1996;271:994-996; PMID:8584938. doi:10.1126/science.271.5251.994.
  • Banks AS, Davis SM, Bates SH, Myers MG Jr Activation of downstream signals by the long form of the leptin receptor. J Biol Chem. 2000;275:14563-14572; PMID:10799542. doi:10.1074/jbc.275.19.14563.
  • Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature. 2003;421:856-859; PMID:12594516. doi:10.1038/nature01388.
  • Gao Q, Wolfgang MJ, Neschen S, Morino K, Horvath TL, Shulman GI, Fu XY. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc Natl Acad Sci USA. 2004;101:4661-4666; PMID:15070774. doi:10.1073/pnas.0303992101.
  • Hekerman P, Zeidler J, Bamberg-Lemper S, Knobelspies H, Lavens D, Tavernier J, Joost HG, Becker W. Pleiotropy of leptin receptor signalling is defined by distinct roles of the intracellular tyrosines. FEBS J. 2005;272:109-119; PMID:15634336. doi:10.1111/j.1432-1033.2004.04391.x.
  • Furigo IC, Ramos-Lobo AM, Frazao R, Donato J Jr Brain STAT5 signaling and behavioral control. Mol Cell Endocrinol. 2016;438:70-76; PMID:27118133. doi:10.1016/j.mce.2016.04.019.
  • Patterson CM, Villanueva EC, Greenwald-Yarnell M, Rajala M, Gonzalez IE, Saini N, Jones J, Myers MG Jr Leptin action via LepR-b Tyr1077 contributes to the control of energy balance and female reproduction. Mol Metab. 2012;1:61-69; PMID:24024119. doi:10.1016/j.molmet.2012.05.001.
  • Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG, Myers MG Jr, Seeley RJ, Schwartz MW. Insulin activation of Phosphatidylinositol 3-Kinase in the hypothalamic arcuate nucleus: a key mediator of Insulin-Induced anorexia. Diabetes. 2003;52:227-231; PMID:12540590. doi:10.2337/diabetes.52.2.227.
  • Donato J Jr, Frazão R, Elias CF. The PI3K signaling pathway mediates the biological effects of leptin. Arq Bras Endocrinol Metabol. 2010;54:591-602; PMID:21085763. doi:10.1590/S0004-27302010000700002.
  • Diano S, Kalra SP, Sakamoto H, Horvath TL. Leptin receptors in estrogen receptor-containing neurons of the female rat hypothalamus. Brain Res. 1998;812:256-259; PMID:9813356. doi:10.1016/S0006-8993(98)00936-6.
  • Roy AF, Benomar Y, Bailleux V, Vacher CM, Aubourg A, Gertler A, Djiane J, Taouis M. Lack of cross-desensitization between leptin and prolactin signaling pathways despite the induction of suppressor of cytokine signaling 3 and PTP-1B. J Endocrinol. 2007;195:341-350; PMID:17951545. doi:10.1677/JOE-07-0321.
  • Mercer JG, Hoggard N, Williams LM, Lawrence CB, Hannah LT, Trayhurn P. Localization of leptin receptor mRNA and the long form splice variant (Ob-Rb) in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS Lett. 1996;387:113-116; PMID:8674530. doi:10.1016/0014-5793(96)00473-5.
  • Schwartz MW, Seeley RJ, Campfield LA, Burn P, Baskin DG. Identification of targets of leptin action in rat hypothalamus. J Clin Invest. 1996;98:1101-1106; PMID:8787671. doi:10.1172/JCI118891.
  • Cheung CC, Clifton DK, Steiner RA. Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology. 1997;138:4489-4492; PMID:9322969. doi:10.1210/endo.138.10.5570.
  • Fei H, Okano HJ, Li C, Lee GH, Zhao C, Darnell R, Friedman JM. Anatomic localization of alternatively spliced leptin receptors (Ob-R) in mouse brain and other tissues. Proc Natl Acad Sci USA. 1997;94:7001-7005; PMID:9192681. doi:10.1073/pnas.94.13.7001.
  • Guan XM, Hess JF, Yu H, Hey PJ, van der Ploeg LH. Differential expression of mRNA for leptin receptor isoforms in the rat brain. Mol Cell Endocrinol. 1997;133:1-7; PMID:9359467. doi:10.1016/S0303-7207(97)00138-X.
  • Bjorbaek C, Elmquist JK, Michl P, Ahima RS, van Bueren A, McCall AL, Flier JS. Expression of leptin receptor isoforms in rat brain microvessels. Endocrinology. 1998;139:3485-3491; PMID:9681499. doi:10.1210/endo.139.8.6154.
  • Hubschle T, Thom E, Watson A, Roth J, Klaus S, Meyerhof W. Leptin-induced nuclear translocation of STAT3 immunoreactivity in hypothalamic nuclei involved in body weight regulation. J Neurosci. 2001;21:2413-2424; PMID:11264315..
  • Hosoi T, Kawagishi T, Okuma Y, Tanaka J, Nomura Y. Brain stem is a direct target for leptin's action in the central nervous system. Endocrinology. 2002;143:3498-3504; PMID:12193563. doi:10.1210/en.2002-220077.
  • Munzberg H, Huo L, Nillni EA, Hollenberg AN, Bjorbaek C. Role of signal transducer and activator of transcription 3 in regulation of hypothalamic proopiomelanocortin gene expression by leptin. Endocrinology. 2003;144:2121-2131; PMID:12697721. doi:10.1210/en.2002-221037.
  • Carpenter LR, Farruggella TJ, Symes A, Karow ML, Yancopoulos GD, Stahl N. Enhancing leptin response by preventing SH2-containing phosphatase 2 interaction with Ob receptor. Proc Natl Acad Sci USA. 1998;95:6061-6066; PMID:9600917. doi:10.1073/pnas.95.11.6061.
  • El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest. 2000;105:1827-1832; PMID:10862798. doi:10.1172/JCI9842.
  • Münzberg H, Flier JS, Bjørbæk C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology. 2004;145:4880-4889; PMID:15271881. doi:10.1210/en.2004-0726.
  • Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, Haj F, Wang Y, Minokoshi Y, Kim YB, Elmquist JK, Tartaglia LA, Kahn BB, et al. PTP1B regulates leptin signal transduction in vivo. Dev Cell. 2002;2:489-495; PMID:11970898. doi:10.1016/S1534-5807(02)00148-X.
  • Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK, Friedman JM. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269:543-546; PMID:7624777. doi:10.1126/science.7624777.
  • Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292-295; PMID:8532024. doi:10.1056/NEJM199602013340503.
  • Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O'Rahilly S. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med. 1999;341:879-884; PMID:10486419. doi:10.1056/NEJM199909163411204.
  • Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110:1093-1103; PMID:12393845. doi:10.1172/JCI0215693.
  • Pelleymounter MA, Cullen MJ, Baker MB, Hecht R, Winters D, Boone T, Collins F. Effects of the obese gene product on body weight regulation in ob/ob mice. Science. 1995;269:540-543; PMID:7624776. doi:10.1126/science.7624776.
  • Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism. 2015;64:24-34; PMID:25199978. doi:10.1016/j.metabol.2014.08.004.
  • Zeltser LM. Developmental influences on circuits programming susceptibility to obesity. Front Neuroendocrinol. 2015;39:17-27; PMID:26206662. doi:10.1016/j.yfrne.2015.07.002.
  • Khan SM, Hamnvik OP, Brinkoetter M, Mantzoros CS. Leptin as a modulator of neuroendocrine function in humans. Yonsei Med J. 2012;53:671-679; PMID:22665330. doi:10.3349/ymj.2012.53.4.671.
  • Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, Flier JS. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382:250-252; PMID:8717038. doi:10.1038/382250a0.
  • Boden G, Chen X, Mozzoli M, Ryan I. Effect of fasting on serum leptin in normal human subjects. J Clin Endocrinol Metab. 1996;81:3419-3423; PMID:8784108. doi:10.1210/jcem.81.9.8784108.
  • Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest. 2003;111:1409-1421; PMID:12727933. doi:10.1172/JCI200317490.
  • Leibel RL. The role of leptin in the control of body weight. Nutr Rev. 2002;60:S15-S19; discussion S68–84, 5–7; PMID:12403079. doi:10.1301/002966402320634788.
  • Campfield LA, Smith FJ, Guisez Y, Devos R, Burn P. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science. 1995;269:546-549; PMID:7624778. doi:10.1126/science.7624778.
  • Halaas JL, Boozer C, Blair-West J, Fidahusein N, Denton DA, Friedman JM. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci USA. 1997;94:8878-8883; PMID:9238071. doi:10.1073/pnas.94.16.8878.
  • Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O'Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA. 2004;101:4531-4536; PMID:15070752. doi:10.1073/pnas.0308767101.
  • Niswender KD, Morton GJ, Stearns WH, Rhodes CJ, Myers MG Jr, Schwartz MW. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature. 2001;413:794-795; PMID:11677594. doi:10.1038/35101657.
  • Hill JW, Williams KW, Ye C, Luo J, Balthasar N, Coppari R, Cowley MA, Cantley LC, Lowell BB, Elmquist JK. Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice. J Clin Invest. 2008;118:1796-1805; PMID:18382766. doi:10.1172/JCI32964.
  • Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, Tang V, Liu SM, Ludwig T, Chua SC Jr, et al. The hypothalamic arcuate nucleus: a key site for mediating leptin's effects on glucose homeostasis and locomotor activity. Cell Metab. 2005;1:63-72; PMID:16054045. doi:10.1016/j.cmet.2004.12.004.
  • Elias CF, Lee C, Kelly J, Aschkenasi C, Ahima RS, Couceyro PR, Kuhar MJ, Saper CB, Elmquist JK. Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron. 1998;21:1375-1385; PMID:9883730. doi:10.1016/S0896-6273(00)80656-X.
  • Schwartz MW, Seeley RJ, Woods SC, Weigle DS, Campfield LA, Burn P, Baskin DG. Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes. 1997;46:2119-2123; PMID:9392508. doi:10.2337/diab.46.12.2119.
  • Thornton JE, Cheung CC, Clifton DK, Steiner RA. Regulation of hypothalamic proopiomelanocortin mRNA by leptin in ob/ob mice. Endocrinology. 1997;138:5063-5066; PMID:9348241. doi:10.1210/endo.138.11.5651.
  • Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL, Cone RD, Low MJ. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature. 2001;411:480-484; PMID:11373681. doi:10.1038/35078085.
  • Edwards CM, Abbott CR, Sunter D, Kim M, Dakin CL, Murphy KG, Abusnana S, Taheri S, Rossi M, Bloom SR. Cocaine- and amphetamine-regulated transcript, glucagon-like peptide-1 and corticotrophin releasing factor inhibit feeding via agouti-related protein independent pathways in the rat. Brain Res. 2000;866:128-134; PMID:10825488. doi:10.1016/S0006-8993(00)02257-5.
  • Rossi M, Kim MS, Morgan DG, Small CJ, Edwards CM, Sunter D, Abusnana S, Goldstone AP, Russell SH, Stanley SA, et al. A C-terminal fragment of agouti-related protein increases feeding and antagonizes the effect of alpha-melanocyte stimulating hormone in vivo. Endocrinology. 1998;139:4428-4431; PMID:9751529. doi:10.1210/endo.139.10.6332.
  • Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88:131-141; PMID:9019399. doi:10.1016/S0092-8674(00)81865-6.
  • Pedroso JA, Silveira MA, Lima LB, Furigo IC, Zampieri TT, Ramos-Lobo AM, Buonfiglio DC, Teixeira PD, Frazão R, Donato J Jr. Changes in leptin signaling by SOCS3 modulate fasting-induced hyperphagia and weight regain in mice. Endocrinology. 2016;157:3901-3914; PMID:27471877. doi:10.1210/en.2016-1038.
  • Hahn TM, Breininger JF, Baskin DG, Schwartz MW. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat Neurosci. 1998;1:271-272; PMID:10195157. doi:10.1038/1082.
  • Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, Maratos-Flier E, Roth BL, Lowell BB. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest. 2011;121:1424-1428; PMID:21364278. doi:10.1172/JCI46229.
  • Gropp E, Shanabrough M, Borok E, Xu AW, Janoschek R, Buch T, Plum L, Balthasar N, Hampel B, Waisman A, et al. Agouti-related peptide-expressing neurons are mandatory for feeding. Nat Neurosci. 2005;8:1289-1291; PMID:16158063. doi:10.1038/nn1548.
  • Clark JT, Kalra PS, Kalra SP. Neuropeptide Y stimulates feeding but inhibits sexual behavior in rats. Endocrinology. 1985;117:2435-2442; PMID:3840737. doi:10.1210/endo-117-6-2435.
  • Ollmann MM, Wilson BD, Yang YK, Kerns JA, Chen Y, Gantz I, Barsh GS. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science. 1997;278:135-138; PMID:9311920. doi:10.1126/science.278.5335.135.
  • Wu Q, Boyle MP, Palmiter RD. Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell. 2009;137:1225-1234; PMID:19563755. doi:10.1016/j.cell.2009.04.022.
  • Tong Q, Ye C-P, Jones JE, Elmquist JK, Lowell BB. Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat Neurosci. 2008;11:998-1000; PMID:19160495. doi:10.1038/nn.2167.
  • Hagan MM, Rushing PA, Schwartz MW, Yagaloff KA, Burn P, Woods SC, Seeley RJ. Role of the CNS melanocortin system in the response to overfeeding. J Neurosci. 1999;19:2362-2367; PMID:10066286..
  • Balthasar N, Coppari R, McMinn J, Liu SM, Lee CE, Tang V, Kenny CD, McGovern RA, Chua SC Jr, Elmquist JK, et al. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron. 2004;42:983-991; PMID:15207242. doi:10.1016/j.neuron.2004.06.004.
  • van de Wall E, Leshan R, Xu AW, Balthasar N, Coppari R, Liu SM, Jo YH, MacKenzie RG, Allison DB, Dun NJ, et al. Collective and individual functions of leptin receptor modulated neurons controlling metabolism and ingestion. Endocrinology. 2008;149:1773-1785; PMID:18162515. doi:10.1210/en.2007-1132.
  • Luquet S, Perez FA, Hnasko TS, Palmiter RD. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science. 2005;310:683-685; PMID:16254186. doi:10.1126/science.1115524.
  • Balthasar N, Dalgaard LT, Lee CE, Yu J, Funahashi H, Williams T, Ferreira M, Tang V, McGovern RA, Kenny CD, et al. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell. 2005;123:493-505; PMID:16269339. doi:10.1016/j.cell.2005.08.035.
  • Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004;304:108-110; PMID:15064420. doi:10.1126/science.1095004.
  • Lima LB, Metzger M, Furigo IC, Donato J Jr Leptin receptor-positive and leptin receptor-negative proopiomelanocortin neurons innervate an identical set of brain structures. Brain Res. 2016;1646:366-376; PMID:27321158. doi:10.1016/j.brainres.2016.06.024.
  • Takahashi KA, Cone RD. Fasting induces a large, leptin-dependent increase in the intrinsic action potential frequency of orexigenic arcuate nucleus neuropeptide Y/Agouti-related protein neurons. Endocrinology. 2005;146:1043-1047; PMID:15591135. doi:10.1210/en.2004-1397.
  • Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa K, Matsukura S. A role for ghrelin in the central regulation of feeding. Nature. 2001;409:194-198; PMID:11196643. doi:10.1038/35051587.
  • Krashes MJ, Shah BP, Madara JC, Olson DP, Strochlic DE, Garfield AS, Vong L, Pei H, Watabe-Uchida M, Uchida N, et al. An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature. 2014;507:238-2342; PMID:24487620. doi:10.1038/nature12956.
  • Liu T, Kong D, Shah Bhavik P, Ye C, Koda S, Saunders A, Ding JB, Yang Z, Sabatini BL, Lowell BB. Fasting activation of AgRP neurons requires NMDA receptors and involves spinogenesis and increased excitatory tone. Neuron. 2012;73:511-522; PMID:22325203. doi:10.1016/j.neuron.2011.11.027.
  • Wu Q, Clark MS, Palmiter RD. Deciphering a neuronal circuit that mediates appetite. Nature. 2012;483:594-597; PMID:22419158. doi:10.1038/nature10899.
  • Carter ME, Soden ME, Zweifel LS, Palmiter RD. Genetic identification of a neural circuit that suppresses appetite. Nature. 2013;503:111-114; PMID:24121436. doi:10.1038/nature12596.
  • Elias CF, Aschkenasi C, Lee C, Kelly J, Ahima RS, Bjorbaek C, Flier JS, Saper CB, Elmquist JK. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron. 1999;23:775-786; PMID:10482243. doi:10.1016/S0896-6273(01)80035-0.
  • Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E. Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature. 1998;396:670-674; PMID:9872314. doi:10.1038/25341.
  • Leinninger GM, Opland DM, Jo YH, Faouzi M, Christensen L, Cappellucci LA, Rhodes CJ, Gnegy ME, Becker JB, Pothos EN, et al. Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance. Cell Metab. 2011;14:313-323; PMID:21907138. doi:10.1016/j.cmet.2011.06.016.
  • Goforth PB, Leinninger GM, Patterson CM, Satin LS, Myers MG Jr Leptin acts via lateral hypothalamic area neurotensin neurons to inhibit orexin neurons by multiple GABA-independent mechanisms. J Neurosci. 2014;34:11405-11415; PMID:25143620. doi:10.1523/JNEUROSCI.5167-13.2014.
  • Opland D, Sutton A, Woodworth H, Brown J, Bugescu R, Garcia A, Christensen L, Rhodes C, Myers M Jr, Leinninger G. Loss of neurotensin receptor-1 disrupts the control of the mesolimbic dopamine system by leptin and promotes hedonic feeding and obesity. Mol Metab. 2013;2:423-434; PMID:24327958. doi:10.1016/j.molmet.2013.07.008.
  • Yu S, Qualls-Creekmore E, Rezai-Zadeh K, Jiang Y, Berthoud HR, Morrison CD, Derbenev AV, Zsombok A, Münzberg H. Glutamatergic preoptic area neurons that express leptin receptors drive temperature-dependent body weight homeostasis. J Neurosci. 2016;36:5034-5046; PMID:27147656. doi:10.1523/JNEUROSCI.0213-16.2016.
  • Hommel JD, Trinko R, Sears RM, Georgescu D, Liu Z-W, Gao X-B, Thurmon JJ, Marinelli M, DiLeone RJ. Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron. 2006;51:801-810; PMID:16982424. doi:10.1016/j.neuron.2006.08.023.
  • Fulton S, Pissios P, Manchon Ramon P, Stiles L, Frank L, Pothos EN, Maratos-Flier E, Flier JS. Leptin regulation of the mesoaccumbens dopamine pathway. Neuron. 2006;51:811-822; PMID:16982425. doi:10.1016/j.neuron.2006.09.006.
  • Xu L, Scheenen WJJM, Leshan RL, Patterson CM, Elias CF, Bouwhuis S, Roubos EW, Myers MG Jr, Kozicz T. Leptin signaling modulates the activity of urocortin 1 neurons in the mouse nonpreganglionic edinger-westphal nucleus. Endocrinology. 2011;152:979-988; PMID:21209012. doi:10.1210/en.2010-1143.
  • Scott MM, Lachey JL, Sternson SM, Lee CE, Elias CF, Friedman JM, Elmquist JK. Leptin targets in the mouse brain. J Comp Neurol. 2009;514:518-532; PMID:19350671. doi:10.1002/cne.22025.
  • Donato J Jr, Frazao R, Fukuda M, Vianna CR, Elias CF. Leptin induces phosphorylation of neuronal nitric oxide synthase in defined hypothalamic neurons. Endocrinology. 2010;151:5415-5427; PMID:20881244. doi:10.1210/en.2010-0651.
  • Nagaishi VS, Cardinali LI, Zampieri TT, Furigo IC, Metzger M, Donato J Jr Possible crosstalk between leptin and prolactin during pregnancy. Neuroscience. 2014;259:71-83; PMID:24316468. doi:10.1016/j.neuroscience.2013.11.050.
  • Garfield AS, Patterson C, Skora S, Gribble FM, Reimann F, Evans ML, Myers MG Jr, Heisler LK. Neurochemical characterization of body weight-regulating leptin receptor neurons in the nucleus of the solitary tract. Endocrinology. 2012;153:4600-4607; PMID:22869346. doi:10.1210/en.2012-1282.
  • Scott MM, Williams KW, Rossi J, Lee CE, Elmquist JK. Leptin receptor expression in hindbrain Glp-1 neurons regulates food intake and energy balance in mice. J Clin Invest. 2011;121:2413-2421; PMID:21606595. doi:10.1172/JCI43703.
  • Hayes MR, Skibicka KP, Leichner TM, Guarnieri DJ, DiLeone RJ, Bence KK, Grill HJ. Endogenous leptin signaling in the caudal nucleus tractus solitarius and area postrema is required for energy balance regulation. Cell Metab. 2010;11:77-83; PMID:20074530. doi:10.1016/j.cmet.2009.10.009.
  • Kim JG, Suyama S, Koch M, Jin S, Argente-Arizon P, Argente J, Liu ZW, Zimmer MR, Jeong JK, Szigeti-Buck K, et al. Leptin signaling in astrocytes regulates hypothalamic neuronal circuits and feeding. Nat Neurosci. 2014;17:908-910; PMID:24880214. doi:10.1038/nn.3725.
  • Trayhurn P, Fuller L. The development of obesity in genetically diabetic-obese (db/db) mice pair-fed with lean siblings. The importance of thermoregulatory thermogenesis. Diabetologia. 1980;19:148-153; PMID:7418967. doi:10.1007/BF00421862.
  • Berglund ED, Vianna CR, Donato J Jr, Kim MH, Chuang JC, Lee CE, Lauzon DA, Lin P, Brule LJ, Scott MM, et al. Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J Clin Invest. 2012;122:1000-1009; PMID:22326958. doi:10.1172/JCI59816.
  • Erickson JC, Hollopeter G, Palmiter RD. Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptide Y. Science. 1996;274:1704-1707; PMID:8939859. doi:10.1126/science.274.5293.1704.
  • Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, Kenny CD, Christiansen LM, White RD, Edelstein EA, et al. Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron. 2006;49:191-203; PMID:16423694. doi:10.1016/j.neuron.2005.12.021.
  • Bingham NC, Anderson KK, Reuter AL, Stallings NR, Parker KL. Selective loss of leptin receptors in the ventromedial hypothalamic nucleus results in increased adiposity and a metabolic syndrome. Endocrinology. 2008;149:2138-2148; PMID:18258679. doi:10.1210/en.2007-1200.
  • Enriori PJ, Sinnayah P, Simonds SE, Garcia Rudaz C, Cowley MA. Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci. 2011;31:12189-12197; PMID:21865462. doi:10.1523/JNEUROSCI.2336-11.2011.
  • Zhang Y, Kerman IA, Laque A, Nguyen P, Faouzi M, Louis GW, Jones JC, Rhodes C, Münzberg H. Leptin-receptor-expressing neurons in the dorsomedial hypothalamus and median preoptic area regulate sympathetic brown adipose tissue circuits. J Neurosci. 2011;31:1873-1884; PMID:21289197. doi:10.1523/JNEUROSCI.3223-10.2011.
  • Rezai-Zadeh K, Yu S, Jiang Y, Laque A, Schwartzenburg C, Morrison CD, Derbenev AV, Zsombok A, Münzberg H. Leptin receptor neurons in the dorsomedial hypothalamus are key regulators of energy expenditure and body weight, but not food intake. Mol Metab. 2014;3:681-693; PMID:25352997. doi:10.1016/j.molmet.2014.07.008.
  • Dodd GT, Worth AA, Nunn N, Korpal AK, Bechtold DA, Allison MB, Myers MG Jr, Statnick MA, Luckman SM. The thermogenic effect of leptin is dependent on a distinct population of prolactin-releasing peptide neurons in the dorsomedial hypothalamus. Cell Metab. 2014;20:639-649; PMID:25176149. doi:10.1016/j.cmet.2014.07.022.
  • Fekete C, Legradi G, Mihaly E, Huang QH, Tatro JB, Rand WM, Emerson CH, Lechan RM. alpha-Melanocyte-stimulating hormone is contained in nerve terminals innervating thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and prevents fasting-induced suppression of prothyrotropin-releasing hormone gene expression. J Neurosci. 2000;20:1550-1558; PMID:10662844.
  • Fekete C, Mihaly E, Luo LG, Kelly J, Clausen JT, Mao Q, Rand WM, Moss LG, Kuhar M, Emerson CH, et al. Association of cocaine- and amphetamine-regulated transcript-immunoreactive elements with thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and its role in the regulation of the hypothalamic-pituitary-thyroid axis during fasting. J Neurosci. 2000;20:9224-9234; PMID:11125000.
  • Kim MS, Small CJ, Stanley SA, Morgan DG, Seal LJ, Kong WM, Edwards CM, Abusnana S, Sunter D, Ghatei MA, et al. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest. 2000;105:1005-1011; PMID:10749579. doi:10.1172/JCI8857.
  • Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bjoorbaek C, Elmquist JK, Flier JS, Hollenberg AN. Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin Invest. 2001;107:111-120; PMID:11134186. doi:10.1172/JCI10741.
  • Morton GJ, Kaiyala KJ, Fisher JD, Ogimoto K, Schwartz MW, Wisse BE. Identification of a physiological role for leptin in the regulation of ambulatory activity and wheel running in mice. Am J Physiol Endocrinol Metab. 2011;300:E392-E401; PMID:21062956. doi:10.1152/ajpendo.00546.2010.
  • Collins S, Kuhn CM, Petro AE, Swick AG, Chrunyk BA, Surwit RS. Role of leptin in fat regulation. Nature. 1996;380:677; PMID:8614460. doi:10.1038/380677a0.
  • Bates SH, Dundon TA, Seifert M, Carlson M, Maratos-Flier E, Myers MG Jr LRb-STAT3 signaling is required for the neuroendocrine regulation of energy expenditure by leptin. Diabetes. 2004;53:3067-3073; PMID:15561935. doi:10.2337/diabetes.53.12.3067.
  • Kaiyala KJ, Ogimoto K, Nelson JT, Muta K, Morton GJ. Physiological role for leptin in the control of thermal conductance. Mol Metab. 2016;5:892-902; PMID:27689002. doi:10.1016/j.molmet.2016.07.005.
  • Fischer AW, Hoefig CS, Abreu-Vieira G, de Jong JM, Petrovic N, Mittag J, Cannon B, Nedergaard J. Leptin raises defended body temperature without activating thermogenesis. Cell Rep. 2016;14:1621-1631; PMID:26876182. doi:10.1016/j.celrep.2016.01.041.
  • Heaton GM, Wagenvoord RJ, Kemp A Jr, Nicholls DG. Brown-adipose-tissue mitochondria: photoaffinity labelling of the regulatory site of energy dissipation. Eur J Biochem. 1978;82:515-521; PMID:624284. doi:10.1111/j.1432-1033.1978.tb12045.x.
  • Bartness TJ, Vaughan CH, Song CK. Sympathetic and sensory innervation of brown adipose tissue. Int J Obes (Lond). 2010;34(Suppl 1):S36-S42; PMID:20935665. doi:10.1038/ijo.2010.182.
  • Scarpace PJ, Matheny M, Pollock BH, Tumer N. Leptin increases uncoupling protein expression and energy expenditure. Am J Physiol. 1997;273:E226-E230; PMID:9252501.
  • van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, Schrauwen P, Teule GJ. Cold-activated brown adipose tissue in healthy men. N Engl J Med. 2009;360:1500-1508; PMID:19357405. doi:10.1056/NEJMoa0808718.
  • Dodd GT, Decherf S, Loh K, Simonds SE, Wiede F, Balland E, Merry TL, Münzberg H, Zhang ZY, Kahn BB, et al. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell. 2015;160:88-104; PMID:25594176. doi:10.1016/j.cell.2014.12.022.
  • Chong AC, Greendyk RA, Zeltser LM. Distinct networks of leptin- and insulin-sensing neurons regulate thermogenic responses to nutritional and cold challenges. Diabetes. 2015;64:137-146; PMID:25125486. doi:10.2337/db14-0567.
  • Geiser F. Metabolic rate and body temperature reduction during hibernation and daily torpor. Annu Rev Physiol. 2004;66:239-274; PMID:14977403. doi:10.1146/annurev.physiol.66.032102.115105.
  • Hudson JW, Scott IM. Daily torpor in the laboratory mouse, Mus-Musculus Var Albino. Physiol Zool. 1979;52:205-218. doi:10.1086/physzool.52.2.30152564.
  • Solymar M, Petervari E, Balasko M, Szelenyi Z. The onset of daily torpor is regulated by the same low body mass in lean mice and in mice with diet-induced obesity. Temperature. 2015;2:129-134. doi:10.1080/23328940.2015.1014250.
  • Faggioni R, Fantuzzi G, Fuller J, Dinarello CA, Feingold KR, Grunfeld C. IL-1 beta mediates leptin induction during inflammation. Am J Physiol. 1998;274:R204-R208; PMID:9458919.
  • Grunfeld C, Zhao C, Fuller J, Pollack A, Moser A, Friedman J, Feingold KR. Endotoxin and cytokines induce expression of leptin, the ob gene product, in hamsters. J Clin Invest. 1996;97:2152-2157; PMID:8621806. doi:10.1172/JCI118653.
  • Sarraf P, Frederich RC, Turner EM, Ma G, Jaskowiak NT, Rivet DJ 3rd, Flier JS, Lowell BB, Fraker DL, Alexander HR. Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia. J Exp Med. 1997;185:171-175; PMID:8996253. doi:10.1084/jem.185.1.171.
  • Finck BN, Kelley KW, Dantzer R, Johnson RW. In vivo and in vitro evidence for the involvement of tumor necrosis factor-alpha in the induction of leptin by lipopolysaccharide. Endocrinology. 1998;139:2278-2283; PMID:9564834. doi:10.1210/endo.139.5.6012.
  • American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med. 1992;20:864-874; PMID:1597042. doi:10.1097/00003246-199206000-00025.
  • Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G. 2001 SCCM/ESICM/ACCP/ATS/SIS International sepsis definitions conference. Crit Care Med. 2003;31:1250-1256; PMID:12682500. doi:10.1097/01.CCM.0000050454.01978.3B.
  • Clemmer TP, Fisher CJ Jr, Bone RC, Slotman GJ, Metz CA, Thomas FO. Hypothermia in the sepsis syndrome and clinical outcome. The Methylprednisolone Severe Sepsis Study Group. Crit Care Med. 1992;20:1395-1401; PMID:1395659. doi:10.1097/00003246-199210000-00006.
  • Rudaya AY, Steiner AA, Robbins JR, Dragic AS, Romanovsky AA. Thermoregulatory responses to lipopolysaccharide in the mouse: dependence on the dose and ambient temperature. Am J Physiol Regul Integr Comp Physiol. 2005;289:R1244-R1252; PMID:16081879. doi:10.1152/ajpregu.00370.2005.
  • Steiner AA, Chakravarty S, Robbins JR, Dragic AS, Pan J, Herkenham M, Romanovsky AA. Thermoregulatory responses of rats to conventional preparations of lipopolysaccharide are caused by lipopolysaccharide per se – not by lipoprotein contaminants. Am J Physiol Regul Integr Comp Physiol. 2005;289:R348-R352; PMID:15860647. doi:10.1152/ajpregu.00223.2005.
  • Romanovsky AA, Szekely M. Fever and hypothermia: two adaptive thermoregulatory responses to systemic inflammation. Med Hypotheses. 1998;50:219-226; PMID:9578327. doi:10.1016/S0306-9877(98)90022-6.
  • Steiner AA, Dogan MD, Ivanov AI, Patel S, Rudaya AY, Jennings DH, Orchinik M, Pace TW, O'connor KA, Watkins LR, et al. A new function of the leptin receptor: mediation of the recovery from lipopolysaccharide-induced hypothermia. FASEB J. 2004;18:1949-1951; PMID:15388670. doi:10.1096/fj.04-2295fje.
  • Faggioni R, Fantuzzi G, Gabay C, Moser A, Dinarello CA, Feingold KR, Grunfeld C. Leptin deficiency enhances sensitivity to endotoxin-induced lethality. Am J Physiol. 1999;276:R136-R142; PMID:9887187.
  • Bik W, Wolinska-Witort E, Chmielowska M, Rusiecka-Kuczalek E, Baranowska B. Does leptin modulate immune and endocrine response in the time of LPS-induced acute inflammation?. Neuro Endocrinol Lett. 2001;22:208-214; PMID:11449193.
  • Faggioni R, Moser A, Feingold KR, Grunfeld C. Reduced leptin levels in starvation increase susceptibility to endotoxic shock. Am J Pathol. 2000;156:1781-1787; PMID:10793089. doi:10.1016/S0002-9440(10)65049-3.
  • Molnar D, Milner RD. Metabolic and hormonal response to endotoxin fever in fed and starved one-week rabbits. Biol Neonate. 1983;44:309-314; PMID:6357299. doi:10.1159/000241732.
  • Shido O, Nagasaka T, Watanabe T. Blunted febrile response to intravenous endotoxin in starved rats. J Appl Physiol. 1989;67:963-969; PMID:2793726.
  • Inoue W, Luheshi GN. Acute starvation alters lipopolysaccharide-induced fever in leptin-dependent and -independent mechanisms in rats. Am J Physiol Regul Integr Comp Physiol. 2010;299:R1709-R1719; PMID:20943858. doi:10.1152/ajpregu.00567.2010.
  • Steiner AA, Romanovsky AA. Leptin: at the crossroads of energy balance and systemic inflammation. Prog Lipid Res. 2007;46:89-107; PMID:17275915. doi:10.1016/j.plipres.2006.11.001.
  • Romanovsky AA, Almeida MC, Aronoff DM, Ivanov AI, Konsman JP, Steiner AA, Turek VF. Fever and hypothermia in systemic inflammation: recent discoveries and revisions. Front Biosci. 2005;10:2193-2216; PMID:15970487. doi:10.2741/1690.
  • Pohl J, Woodside B, Luheshi GN. Leptin modulates the late fever response to LPS in diet-induced obese animals. Brain Behav Immun. 2014;42:41-47; PMID:25108212. doi:10.1016/j.bbi.2014.07.017.
  • Koenig S, Luheshi GN, Wenz T, Gerstberger R, Roth J, Rummel C. Leptin is involved in age-dependent changes in response to systemic inflammation in the rat. Brain Behav Immun. 2014;36:128-138; PMID:24513873. doi:10.1016/j.bbi.2013.10.019.
  • Timmerman RJ, Thompson J, Noordzij HM, van der Meer JW. Psychogenic periodic fever. Nethe J Med. 1992;41:158-160; PMID:1470287.
  • Lkhagvasuren B, Nakamura Y, Oka T, Sudo N, Nakamura K. Social defeat stress induces hyperthermia through activation of thermoregulatory sympathetic premotor neurons in the medullary raphe region. Eur J Neurosci. 2011;34:1442-1452; PMID:21978215. doi:10.1111/j.1460-9568.2011.07863.x.
  • Nakamura K. Neural circuit for psychological stress-induced hyperthermia. Temperature. 2015;2:352-361; PMID:27227049. doi:10.1080/23328940.2015.1070944.
  • Simonds SE, Pryor JT, Ravussin E, Greenway FL, Dileone R, Allen AM, Bassi J, Elmquist JK, Keogh JM, Henning E, et al. Leptin mediates the increase in blood pressure associated with obesity. Cell. 2014;159:1404-1416; PMID:25480301. doi:10.1016/j.cell.2014.10.058.
  • Tallam LS, Stec DE, Willis MA, da Silva AA, Hall JE. Melanocortin-4 receptor-deficient mice are not hypertensive or salt-sensitive despite obesity, hyperinsulinemia, and hyperleptinemia. Hypertension. 2005;46:326-332; PMID:16027245. doi:10.1161/01.HYP.0000175474.99326.bf.
  • Sohn J-W, Harris Louise E, Berglund Eric D, Liu T, Vong L, Lowell Bradford B, Balthasar N, Williams KW, Elmquist JK. Melanocortin 4 receptors reciprocally regulate sympathetic and parasympathetic preganglionic neurons. Cell. 2013;152:612-619; PMID:23374353. doi:10.1016/j.cell.2012.12.022.
  • Rossi J, Balthasar N, Olson D, Scott M, Berglund E, Lee CE, Choi MJ, Lauzon D, Lowell BB, Elmquist JK. Melanocortin-4 receptors expressed by cholinergic neurons regulate energy balance and glucose homeostasis. Cell Metab. 2011;13:195-204; PMID:21284986. doi:10.1016/j.cmet.2011.01.010.
  • Eikelis N, Schlaich M, Aggarwal A, Kaye D, Esler M. Interactions between leptin and the human sympathetic nervous system. Hypertension. 2003;41:1072-1079; PMID:12668587. doi:10.1161/01.HYP.0000066289.17754.49.
  • Pedroso JA, Buonfiglio DC, Cardinali LI, Furigo IC, Ramos-Lobo AM, Tirapegui J, Elias CF, Donato J Jr Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol Metab. 2014;3:608-618; PMID:25161884. doi:10.1016/j.molmet.2014.06.001.
  • Huo L, Gamber K, Greeley S, Silva J, Huntoon N, Leng XH, Bjørbaek C. Leptin-dependent control of glucose balance and locomotor activity by POMC neurons. Cell Metab. 2009;9:537-547; PMID:19490908. doi:10.1016/j.cmet.2009.05.003.
  • Zhang R, Dhillon H, Yin H, Yoshimura A, Lowell BB, Maratos-Flier E, Flier JS. Selective inactivation of Socs3 in SF1 neurons improves glucose homeostasis without affecting body weight. Endocrinology. 2008;149:5654-5661; PMID:18669597. doi:10.1210/en.2008-0805.
  • Meek TH, Nelson JT, Matsen ME, Dorfman MD, Guyenet SJ, Damian V, Allison MB, Scarlett JM, Nguyen HT, Thaler JP, et al. Functional identification of a neurocircuit regulating blood glucose. Proc Natl Acad Sci USA. 2016;113:E2073-E2082; PMID:27001850. doi:10.1073/pnas.1521160113.
  • Tong Q, Ye C, McCrimmon RJ, Dhillon H, Choi B, Kramer MD, Yu J, Yang Z, Christiansen LM, Lee CE, et al. Synaptic glutamate release by ventromedial hypothalamic neurons is part of the neurocircuitry that prevents hypoglycemia. Cell Metab. 2007;5:383-393; PMID:17488640. doi:10.1016/j.cmet.2007.04.001.
  • Garfield AS, Shah BP, Madara JC, Burke LK, Patterson CM, Flak J, Neve RL, Evans ML, Lowell BB, Myers MG Jr, et al. A parabrachial-hypothalamic cholecystokinin neurocircuit controls counterregulatory responses to hypoglycemia. Cell Metab. 2014;20:1030-1037; PMID:25470549. doi:10.1016/j.cmet.2014.11.006.
  • Flak JN, Patterson CM, Garfield AS, D'Agostino G, Goforth PB, Sutton AK, Malec PA, Wong JM, Germani M, Jones JC, et al. Leptin-inhibited PBN neurons enhance responses to hypoglycemia in negative energy balance. Nat Neurosci. 2014;17:1744-1750; PMID:25383904. doi:10.1038/nn.3861.
  • Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351:987-997; PMID:15342807. doi:10.1056/NEJMoa040388.
  • Bohlen TM, Silveira MA, Zampieri TT, Frazao R, Donato J Jr Fatness rather than leptin sensitivity determines the timing of puberty in female mice. Mol Cell Endocrinol. 2016;423:11-21; PMID:26762764. doi:10.1016/j.mce.2015.12.022.
  • Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, Herbison AE, Grattan DR, Anderson GM. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology. 2009;150:2805-2812; PMID:19179437. doi:10.1210/en.2008-1693.
  • Frazao R, Cravo RM, Donato J Jr, Ratra DV, Clegg DJ, Elmquist JK, Zigman JM, Williams KW, Elias CF. Shift in Kiss1 cell activity requires estrogen receptor alpha. J Neurosci. 2013;33:2807-2820; PMID:23407940. doi:10.1523/JNEUROSCI.1610-12.2013.
  • Smith JT, Acohido BV, Clifton DK, Steiner RA. KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J Neuroendocrinol. 2006;18:298-303; PMID:16503925. doi:10.1111/j.1365-2826.2006.01417.x.
  • Cravo RM, Frazao R, Perello M, Osborne-Lawrence S, Williams KW, Zigman JM, Vianna C, Elias CF. Leptin signaling in Kiss1 neurons arises after pubertal development. PLoS One. 2013;8:e58698; PMID:23505551. doi:10.1371/journal.pone.0058698.
  • Cravo RM, Margatho LO, Osborne-Lawrence S, Donato J Jr, Atkin S, Bookout AL, Rovinsky S, Frazão R, Lee CE, Gautron L, et al. Characterization of Kiss1 neurons using transgenic mouse models. Neuroscience. 2011;173:37-56; PMID:21093546. doi:10.1016/j.neuroscience.2010.11.022.
  • Donato J Jr, Cravo RM, Frazão R, Gautron L, Scott MM, Lachey J, Castro IA, Margatho LO, Lee S, Lee C, et al. Leptin's effect on puberty in mice is relayed by the ventral premammillary nucleus and does not require signaling in Kiss1 neurons. J Clin Invest. 2011;121:355-368; PMID:21183787. doi:10.1172/JCI45106.
  • Donato J Jr, Cravo RM, Frazão R, Elias CF. Hypothalamic sites of leptin action linking metabolism and reproduction. Neuroendocrinology. 2011;93:9-18; PMID:21099209. doi:10.1159/000322472.
  • Donato J Jr, Elias CF. The ventral premammillary nucleus links metabolic cues and reproduction. Front Endocrinol (Lausanne). 2011;2:57; PMID:22649378.
  • Rondini TA, Baddini SP, Sousa LF, Bittencourt JC, Elias CF. Hypothalamic cocaine- and amphetamine-regulated transcript neurons project to areas expressing gonadotropin releasing hormone immunoreactivity and to the anteroventral periventricular nucleus in male and female rats. Neuroscience. 2004;125:735-748; PMID:15099687. doi:10.1016/j.neuroscience.2003.12.045.
  • Leshan RL, Louis GW, Jo Y-H, Rhodes CJ, Munzberg H, Myers MG Jr Direct innervation of GnRH neurons by metabolic- and sexual odorant-sensing leptin receptor neurons in the hypothalamic ventral premammillary nucleus. J Neurosci. 2009;29:3138-3147; PMID:19279251. doi:10.1523/JNEUROSCI.0155-09.2009.
  • Donato J Jr, Lee C, Ratra DV, Franci CR, Canteras NS, Elias CF. Lesions of the ventral premammillary nucleus disrupt the dynamic changes in Kiss1 and GnRH expression characteristic of the proestrus-estrus transition. Neuroscience. 2013;241:67-79; PMID:23518222. doi:10.1016/j.neuroscience.2013.03.013.
  • Donato J Jr, Silva RJ, Sita LV, Lee S, Lee C, Lacchini S, Bittencourt JC, Franci CR, Canteras NS, Elias CF. The ventral premammillary nucleus links fasting-induced changes in leptin levels and coordinated luteinizing hormone secretion. J Neurosci. 2009;29:5240-5250; PMID:19386920. doi:10.1523/JNEUROSCI.0405-09.2009.
  • Bellefontaine N, Chachlaki K, Parkash J, Vanacker C, Colledge W, d'Anglemont de Tassigny X, Garthwaite J, Bouret SG, Prevot V. Leptin-dependent neuronal NO signaling in the preoptic hypothalamus facilitates reproduction. J Clin Invest. 2014;124:2550-2559; PMID:24812663. doi:10.1172/JCI65928.
  • Sheffer-Babila S, Sun Y, Israel DD, Liu SM, Neal-Perry G, Chua SC Jr Agouti-related peptide plays a critical role in leptin's effects on female puberty and reproduction. Am J Physiol Endocrinol Metab. 2013;305:E1512-E1520; PMID:24169048. doi:10.1152/ajpendo.00241.2013.
  • Wu Q, Whiddon BB, Palmiter RD. Ablation of neurons expressing agouti-related protein, but not melanin concentrating hormone, in leptin-deficient mice restores metabolic functions and fertility. Proc Natl Acad Sci USA. 2012;109:3155-3160; PMID:22232663. doi:10.1073/pnas.1120501109.
  • Roa J, Herbison AE. Direct regulation of GnRH neuron excitability by arcuate nucleus POMC and NPY neuron neuropeptides in female mice. Endocrinology. 2012;153:5587-5599; PMID:22948210. doi:10.1210/en.2012-1470.
  • Hill JW, Elias CF, Fukuda M, Williams KW, Berglund ED, Holland WL, Cho YR, Chuang JC, Xu Y, Choi M, et al. Direct insulin and leptin action on pro-opiomelanocortin neurons is required for normal glucose homeostasis and fertility. Cell Metab. 2010;11:286-297; PMID:20374961. doi:10.1016/j.cmet.2010.03.002.
  • Liu J, Perez SM, Zhang W, Lodge DJ, Lu XY. Selective deletion of the leptin receptor in dopamine neurons produces anxiogenic-like behavior and increases dopaminergic activity in amygdala. Mol Psychiatry. 2011;16:1024-1038; PMID:21483433. doi:10.1038/mp.2011.36.
  • Bereiter DA, Jeanrenaud B. Altered neuroanatomical organization in the central nervous system of the genetically obese (ob/ob) mouse. Brain Res. 1979;165:249-260; PMID:421139. doi:10.1016/0006-8993(79)90557-2.
  • Ahima RS, Bjorbaek C, Osei S, Flier JS. Regulation of neuronal and glial proteins by leptin: implications for brain development. Endocrinology. 1999;140:2755-2762; PMID:10342866. doi:10.1210/endo.140.6.6774.
  • Matochik JA, London ED, Yildiz BO, Ozata M, Caglayan S, DePaoli AM, Wong ML, Licinio J. Effect of leptin replacement on brain structure in genetically leptin-deficient adults. J Clin Endocrinol Metab. 2005;90:2851-2854; PMID:15713712. doi:10.1210/jc.2004-1979.
  • Mistry AM, Swick A, Romsos DR. Leptin alters metabolic rates before acquisition of its anorectic effect in developing neonatal mice. Am J Physiol. 1999;277:R742-R747; PMID:10484491.
  • Ishii Y, Bouret SG. Embryonic birthdate of hypothalamic leptin-activated neurons in mice. Endocrinology. 2012;153:3657-3667; PMID:22621961. doi:10.1210/en.2012-1328.
  • Pan W, Hsuchou H, Tu H, Kastin AJ. Developmental changes of leptin receptors in cerebral microvessels: unexpected relation to leptin transport. Endocrinology. 2008;149:877-885; PMID:18039787. doi:10.1210/en.2007-0893.
  • Bouret SG. Crossing the border: developmental regulation of leptin transport to the brain. Endocrinology. 2008;149:875-876; PMID:18292198. doi:10.1210/en.2007-1698.
  • Ahima RS, Prabakaran D, Flier JS. Postnatal leptin surge and regulation of circadian rhythm of leptin by feeding. Implications for energy homeostasis and neuroendocrine function. J Clin Invest. 1998;101:1020-1027; PMID:9486972. doi:10.1172/JCI1176.
  • Markakis EA. Development of the neuroendocrine hypothalamus. Front Neuroendocrinol. 2002;23:257-291; PMID:12127306. doi:10.1016/S0091-3022(02)00003-1.
  • Bouret SG, Draper SJ, Simerly RB. Formation of projection pathways from the arcuate nucleus of the hypothalamus to hypothalamic regions implicated in the neural control of feeding behavior in mice. J Neurosci. 2004;24:2797-2805; PMID:15028773. doi:10.1523/JNEUROSCI.5369-03.2004.
  • Pinto S, Roseberry AG, Liu H, Diano S, Shanabrough M, Cai X, Friedman JM, Horvath TL. Rapid rewiring of arcuate nucleus feeding circuits by leptin. Science. 2004;304:110-115; PMID:15064421. doi:10.1126/science.1089459.
  • O'Malley D, Irving AJ, Harvey J. Leptin-induced dynamic alterations in the actin cytoskeleton mediate the activation and synaptic clustering of BK channels. FASEB J. 2005;19:1917-1919; PMID:16166199. doi:10.1096/fj.05-4166fje.
  • Shanley LJ, O'Malley D, Irving AJ, Ashford ML, Harvey J. Leptin inhibits epileptiform-like activity in rat hippocampal neurones via PI 3-kinase-driven activation of BK channels. J Physiol. 2002;545:933-944; PMID:12482897. doi:10.1113/jphysiol.2002.029488.
  • Ayyildiz M, Yildirim M, Agar E, Baltaci AK. The effect of leptin on penicillin-induced epileptiform activity in rats. Brain Res Bull. 2006;68:374-378; PMID:16377445. doi:10.1016/j.brainresbull.2005.09.012.
  • Li XL, Aou S, Oomura Y, Hori N, Fukunaga K, Hori T. Impairment of long-term potentiation and spatial memory in leptin receptor-deficient rodents. Neuroscience. 2002;113:607-615; PMID:12150780. doi:10.1016/S0306-4522(02)00162-8.
  • Sena A, Sarlieve LL, Rebel G. Brain myelin of genetically obese mice. J Neurol Sci. 1985;68:233-243; PMID:2989440. doi:10.1016/0022-510X(85)90104-2.
  • Winocur G, Greenwood CE, Piroli GG, Grillo CA, Reznikov LR, Reagan LP, McEwen BS. Memory impairment in obese Zucker rats: an investigation of cognitive function in an animal model of insulin resistance and obesity. Behav Neurosci. 2005;119:1389-1395; PMID:16300445. doi:10.1037/0735-7044.119.5.1389.
  • Paz-Filho GJ, Babikian T, Asarnow R, Delibasi T, Esposito K, Erol HK, Wong M-L, Licinio J. Leptin replacement improves cognitive development. PLoS One. 2008;3:e3098; PMID:18769731. doi:10.1371/journal.pone.0003098.
  • Sharma AN, Elased KM, Garrett TL, Lucot JB. Neurobehavioral deficits in db/db diabetic mice. Physiol Behav. 2010;101:381-388; PMID:20637218. doi:10.1016/j.physbeh.2010.07.002.
  • Harvey J. Leptin regulation of neuronal excitability and cognitive function. Curr Opin Pharmacol. 2007;7:643-647; PMID:18024215. doi:10.1016/j.coph.2007.10.006.
  • Lu X-Y, Kim CS, Frazer A, Zhang W. Leptin: a potential novel antidepressant. Proc Natl Acad Sci USA. 2006;103:1593-1598; PMID:16423896. doi:10.1073/pnas.0508901103.
  • Wabitsch M, Funcke JB, Lennerz B, Kuhnle-Krahl U, Lahr G, Debatin KM, Vatter P, Gierschik P, Moepps B, Fischer-Posovszky P. Biologically inactive leptin and early-onset extreme obesity. N Engl J Med. 2015;372:48-54; PMID:25551525. doi:10.1056/NEJMoa1406653.
  • Farooqi S, O'Rahilly S. Genetics of obesity in humans. Endocr Rev. 2006;27:710-718; PMID:17122358. doi:10.1210/er.2006-0040.
  • Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, Lubina JA, Patane J, Self B, Hunt P, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999;282:1568-1575; PMID:10546697. doi:10.1001/jama.282.16.1568.
  • Hukshorn CJ, Saris WH, Westerterp-Plantenga MS, Farid AR, Smith FJ, Campfield LA. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J Clin Endocrinol Metab. 2000;85:4003-4009; PMID:11095423. doi:10.1210/jcem.85.11.6955.
  • Hukshorn CJ, van Dielen FM, Buurman WA, Westerterp-Plantenga MS, Campfield LA, Saris WH. The effect of pegylated recombinant human leptin (PEG-OB) on weight loss and inflammatory status in obese subjects. Int J Obes Relat Metab Disord. 2002;26:504-509; PMID:12075577. doi:10.1038/sj.ijo.0801952.
  • Hukshorn CJ, Menheere PP, Westerterp-Plantenga MS, Saris WH. The effect of pegylated human recombinant leptin (PEG-OB) on neuroendocrine adaptations to semi-starvation in overweight men. Eur J Endocrinol. 2003;148:649-655; PMID:12773137. doi:10.1530/eje.0.1480649.
  • Hukshorn CJ, Westerterp-Plantenga MS, Saris WH. Pegylated human recombinant leptin (PEG-OB) causes additional weight loss in severely energy-restricted, overweight men. Am J Clin Nutr. 2003;77:771-776; PMID:12663271.
  • Lejeune MP, Hukshorn CJ, Saris WH, Westerterp-Plantenga MS. Effect of dietary restraint during and following pegylated recombinant leptin (PEG-OB) treatment of overweight men. Int J Obes Relat Metab Disord. 2003;27:1494-1499; PMID:14634680. doi:10.1038/sj.ijo.0802431.
  • Korner J, Conroy R, Febres G, McMahon DJ, Conwell I, Karmally W, Aronne LJ. Randomized double-blind placebo-controlled study of leptin administration after gastric bypass. Obesity (Silver Spring). 2013;21:951-956; PMID:23512892. doi:10.1002/oby.20433.
  • Shetty GK, Matarese G, Magkos F, Moon HS, Liu X, Brennan AM, Mylvaganam G, Sykoutri D, Depaoli AM, Mantzoros CS. Leptin administration to overweight and obese subjects for 6 months increases free leptin concentrations but does not alter circulating hormones of the thyroid and IGF axes during weight loss induced by a mild hypocaloric diet. Eur J Endocrinol. 2011;165:249-254; PMID:21602313. doi:10.1530/EJE-11-0252.
  • Kissileff HR, Thornton JC, Torres MI, Pavlovich K, Mayer LS, Kalari V, Leibel RL, Rosenbaum M. Leptin reverses declines in satiation in weight-reduced obese humans. Am J Clin Nutr. 2012;95:309-317; PMID:22237063. doi:10.3945/ajcn.111.012385.
  • Rosenbaum M, Sy M, Pavlovich K, Leibel RL, Hirsch J. Leptin reverses weight loss-induced changes in regional neural activity responses to visual food stimuli. J Clin Invest. 2008;118:2583-2591; PMID:18568078. doi:10.1172/JCI35055.
  • Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL. Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature. 1999;401:73-76; PMID:10485707. doi:10.1038/43448.
  • Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, Wagner AJ, DePaoli AM, Reitman ML, Taylor SI, et al. Leptin-replacement therapy for lipodystrophy. N Engl J Med. 2002;346:570-578; PMID:11856796. doi:10.1056/NEJMoa012437.
  • Petersen KF, Oral EA, Dufour S, Befroy D, Ariyan C, Yu C, Cline GW, DePaoli AM, Taylor SI, Gorden P, et al. Leptin reverses insulin resistance and hepatic steatosis in patients with severe lipodystrophy. J Clin Invest. 2002;109:1345-1350; PMID:12021250. doi:10.1172/JCI0215001.
  • Javor ED, Cochran EK, Musso C, Young JR, DePaoli AM, Gorden P. Long-term efficacy of leptin replacement in patients with generalized lipodystrophy. Diabetes. 2005;54:1994-2002; PMID:15983199. doi:10.2337/diabetes.54.7.1994.
  • Sekhar RV, Jahoor F, Iyer D, Guthikonda A, Paranilam J, Elhaj F, Coraza I, Balasubramanyam A. Leptin replacement therapy does not improve the abnormal lipid kinetics of hypoleptinemic patients with HIV-associated lipodystrophy syndrome. Metabolism. 2012;61:1395-1403; PMID:22542724. doi:10.1016/j.metabol.2012.03.013.
  • Magkos F, Brennan A, Sweeney L, Kang ES, Doweiko J, Karchmer AW, Mantzoros CS. Leptin replacement improves postprandial glycemia and insulin sensitivity in human immunodeficiency virus-infected lipoatrophic men treated with pioglitazone: a pilot study. Metabolism. 2011;60:1045-1049; PMID:21081243. doi:10.1016/j.metabol.2010.10.002.
  • Park JY, Chong AY, Cochran EK, Kleiner DE, Haller MJ, Schatz DA, Gorden P. Type 1 diabetes associated with acquired generalized lipodystrophy and insulin resistance: the effect of long-term leptin therapy. J Clin Endocrinol Metab. 2008;93:26-31; PMID:17940115. doi:10.1210/jc.2007-1856.
  • Mittendorfer B, Horowitz JF, DePaoli AM, McCamish MA, Patterson BW, Klein S. Recombinant human leptin treatment does not improve insulin action in obese subjects with type 2 diabetes. Diabetes. 2011;60:1474-1477; PMID:21411512. doi:10.2337/db10-1302.
  • Moon HS, Matarese G, Brennan AM, Chamberland JP, Liu X, Fiorenza CG, Mylvaganam GH, Abanni L, Carbone F, Williams CJ, et al. Efficacy of metreleptin in obese patients with type 2 diabetes: cellular and molecular pathways underlying leptin tolerance. Diabetes. 2011;60:1647-1656; PMID:21617185. doi:10.2337/db10-1791.
  • Wang MY, Chen L, Clark GO, Lee Y, Stevens RD, Ilkayeva OR, Wenner BR, Bain JR, Charron MJ, Newgard CB, et al. Leptin therapy in insulin-deficient type I diabetes. Proc Natl Acad Sci USA. 2010;107:4813-4819; PMID:20194735. doi:10.1073/pnas.0909422107.
  • Fujikawa T, Chuang JC, Sakata I, Ramadori G, Coppari R. Leptin therapy improves insulin-deficient type 1 diabetes by CNS-dependent mechanisms in mice. Proc Natl Acad Sci USA. 2010;107:17391-17396; PMID:20855609. doi:10.1073/pnas.1008025107.
  • Fujikawa T, Berglund ED, Patel VR, Ramadori G, Vianna CR, Vong L, Thorel F, Chera S, Herrera PL, Lowell BB, et al. Leptin engages a hypothalamic neurocircuitry to permit survival in the absence of insulin. Cell Metab. 2013;18:431-444; PMID:24011077. doi:10.1016/j.cmet.2013.08.004.
  • Raskin P, Unger RH. Hyperglucagonemia and its suppression. Importance in the metabolic control of diabetes. N Engl J Med. 1978;299:433-436; PMID:683275. doi:10.1056/NEJM197808312990901.
  • Lee Y, Wang MY, Du XQ, Charron MJ, Unger RH. Glucagon receptor knockout prevents insulin-deficient type 1 diabetes in mice. Diabetes. 2011;60:391-397; PMID:21270251. doi:10.2337/db10-0426.
  • Lee Y, Berglund ED, Wang MY, Fu X, Yu X, Charron MJ, Burgess SC, Unger RH. Metabolic manifestations of insulin deficiency do not occur without glucagon action. Proc Natl Acad Sci USA. 2012;109:14972-14976; PMID:22891336. doi:10.1073/pnas.1205983109.
  • Lee Y, Berglund ED, Yu X, Wang MY, Evans MR, Scherer PE, Holland WL, Charron MJ, Roth MG, Unger RH. Hyperglycemia in rodent models of type 2 diabetes requires insulin-resistant alpha cells. Proc Natl Acad Sci USA. 2014;111:13217-13222; PMID:25157166. doi:10.1073/pnas.1409638111.
  • Chou SH, Chamberland JP, Liu X, Matarese G, Gao C, Stefanakis R, Brinkoetter MT, Gong H, Arampatzi K, Mantzoros CS. Leptin is an effective treatment for hypothalamic amenorrhea. Proc Natl Acad Sci USA. 2011;108:6585-6590; PMID:21464293. doi:10.1073/pnas.1015674108.
  • Sienkiewicz E, Magkos F, Aronis KN, Brinkoetter M, Chamberland JP, Chou S, Arampatzi KM, Gao C, Koniaris A, Mantzoros CS. Long-term metreleptin treatment increases bone mineral density and content at the lumbar spine of lean hypoleptinemic women. Metabolism. 2011;60:1211-1221; PMID:21741057. doi:10.1016/j.metabol.2011.05.016.
  • Van Heek M, Compton DS, France CF, Tedesco RP, Fawzi AB, Graziano MP, Sybertz EJ, Strader CD, Davis HR Jr Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J Clin Invest. 1997;99:385-390; PMID:9022070. doi:10.1172/JCI119171.
  • Ottaway N, Mahbod P, Rivero B, Norman LA, Gertler A, D'Alessio DA, Perez-Tilve D. Diet-induced obese mice retain endogenous leptin action. Cell Metab. 2015;21:877-882; PMID:25980347. doi:10.1016/j.cmet.2015.04.015.
  • Olofsson LE, Unger EK, Cheung CC, Xu AW. Modulation of AgRP-neuronal function by SOCS3 as an initiating event in diet-induced hypothalamic leptin resistance. Proc Natl Acad Sci USA. 2013;110:E697-E706; PMID:23386726. doi:10.1073/pnas.1218284110.
  • Mounzih K, Qiu J, Ewart-Toland A, Chehab FF. Leptin is not necessary for gestation and parturition but regulates maternal nutrition via a leptin resistance state. Endocrinology. 1998;139:5259-5262; PMID:9832467. doi:10.1210/endo.139.12.6523.
  • Ladyman SR, Grattan DR. Suppression of leptin receptor messenger ribonucleic acid and leptin responsiveness in the ventromedial nucleus of the hypothalamus during pregnancy in the rat. Endocrinology. 2005;146:3868-3874; PMID:15905318. doi:10.1210/en.2005-0194.
  • Ladyman SR, Fieldwick DM, Grattan DR. Suppression of leptin-induced hypothalamic JAK/STAT signalling and feeding response during pregnancy in the mouse. Reproduction. 2012;144:83-90; PMID:22580369. doi:10.1530/REP-12-0112.
  • Zampieri TT, Ramos-Lobo AM, Furigo IC, Pedroso JA, Buonfiglio DC, Donato J Jr SOCS3 deficiency in leptin receptor-expressing cells mitigates the development of pregnancy-induced metabolic changes. Mol Metab. 2015;4:237-245; PMID:25737950. doi:10.1016/j.molmet.2014.12.005.
  • Zampieri TT, da Silva TE, de Paula Romeu D, da Silva Torrao A, Donato J Jr SOCS3 expression within leptin receptor-expressing cells regulates food intake and leptin sensitivity but does not affect weight gain in pregnant mice consuming a high-fat diet. Physiol Behav. 2016;157:109-115; PMID:26828039. doi:10.1016/j.physbeh.2016.01.039.
  • Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. Leptin enters the brain by a saturable system independent of insulin. Peptides. 1996;17:305-311; PMID:8801538. doi:10.1016/0196-9781(96)00025-3.
  • Caro JF, Kolaczynski JW, Nyce MR, Ohannesian JP, Opentanova I, Goldman WH, Lynn RB, Zhang PL, Sinha MK, Considine RV. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet. 1996;348:159-161; PMID:8684156. doi:10.1016/S0140-6736(96)03173-X.
  • Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte D Jr Cerebrospinal fluid leptin levels: relationship to plasma levels and to adiposity in humans. Nat Med. 1996;2:589-593; PMID:8616722. doi:10.1038/nm0596-589.
  • Kastin AJ, Pan W, Maness LM, Koletsky RJ, Ernsberger P. Decreased transport of leptin across the blood-brain barrier in rats lacking the short form of the leptin receptor. Peptides. 1999;20:1449-1453; PMID:10698121. doi:10.1016/S0196-9781(99)00156-4.
  • Hileman SM, Tornoe J, Flier JS, Bjorbaek C. Transcellular transport of leptin by the short leptin receptor isoform ObRa in Madin-Darby canine kidney cells. Endocrinology. 2000;141:1955-1961; PMID:10830277. doi:10.1210/endo.141.6.7450.
  • Banks WA, Niehoff ML, Martin D, Farrell CL. Leptin transport across the blood–brain barrier of the Koletsky rat is not mediated by a product of the leptin receptor gene. Brain Res. 2002;950:130-136; PMID:12231237. doi:10.1016/S0006-8993(02)03013-5.
  • Li Z, Ceccarini G, Eisenstein M, Tan K, Friedman JM. Phenotypic effects of an induced mutation of the ObRa isoform of the leptin receptor. Mol Metab. 2013;2:364-375; PMID:24327953. doi:10.1016/j.molmet.2013.07.007.
  • Balland E, Dam J, Langlet F, Caron E, Steculorum S, Messina A, Rasika S, Falluel-Morel A, Anouar Y, Dehouck B, et al. Hypothalamic tanycytes are an ERK-gated conduit for leptin into the brain. Cell Metab. 2014;19:293-301; PMID:24506870. doi:10.1016/j.cmet.2013.12.015.
  • Myers MG Jr, Leibel RL, Seeley RJ, Schwartz MW. Obesity and leptin resistance: distinguishing cause from effect. Trends Endocrinol Metab. 2010;21:643-651; PMID:20846876. doi:10.1016/j.tem.2010.08.002.
  • Ravussin Y, LeDuc CA, Watanabe K, Mueller BR, Skowronski A, Rosenbaum M, Leibel RL. Effects of chronic leptin infusion on subsequent body weight and composition in mice: can body weight set point be reset?. Mol Metab. 2014;3:432-440; PMID:24944902. doi:10.1016/j.molmet.2014.02.003.
  • Krebs DL, Hilton DJ. SOCS proteins: negative regulators of cytokine signaling. Stem Cell. 2001;19:378-387; PMID:11553846. doi:10.1634/stemcells.19-5-378.
  • Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell. 1998;1:619-625; PMID:9660946. doi:10.1016/S1097-2765(00)80062-3.
  • Bjorbaek C, El-Haschimi K, Frantz JD, Flier JS. The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem. 1999;274:30059-30065; PMID:10514492. doi:10.1074/jbc.274.42.30059.
  • Bjornholm M, Munzberg H, Leshan RL, Villanueva EC, Bates SH, Louis GW, Jones JC, Ishida-Takahashi R, Bjørbaek C, Myers MG Jr. Mice lacking inhibitory leptin receptor signals are lean with normal endocrine function. J Clin Invest. 2007;117:1354-1360; PMID:17415414. doi:10.1172/JCI30688.
  • Kievit P, Howard JK, Badman MK, Balthasar N, Coppari R, Mori H, Lee CE, Elmquist JK, Yoshimura A, Flier JS. Enhanced leptin sensitivity and improved glucose homeostasis in mice lacking suppressor of cytokine signaling-3 in POMC-expressing cells. Cell Metab. 2006;4:123-132; PMID:16890540. doi:10.1016/j.cmet.2006.06.010.
  • Briancon N, McNay DE, Maratos-Flier E, Flier JS. Combined neural inactivation of suppressor of cytokine signaling-3 and protein-tyrosine phosphatase-1B reveals additive, synergistic, and factor-specific roles in the regulation of body energy balance. Diabetes. 2010;59:3074-3084; PMID:20876718. doi:10.2337/db10-0481.
  • Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjorbaek C, Flier JS. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med. 2004;10:734-738; PMID:15220914. doi:10.1038/nm1072.
  • Mori H, Hanada R, Hanada T, Aki D, Mashima R, Nishinakamura H, Torisu T, Chien KR, Yasukawa H, Yoshimura A. Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat Med. 2004;10:739-743; PMID:15208705. doi:10.1038/nm1071.
  • Matarazzo V, Schaller F, Nedelec E, Benani A, Penicaud L, Muscatelli F, Moyse E, Bauer S. Inactivation of Socs3 in the hypothalamus enhances the hindbrain response to endogenous satiety signals via oxytocin signaling. J Neurosci. 2012;32:17097-17107; PMID:23197703. doi:10.1523/JNEUROSCI.1669-12.2012.
  • Reed AS, Unger EK, Olofsson LE, Piper ML, Myers MG Jr, Xu AW. Functional role of suppressor of cytokine signaling 3 upregulation in hypothalamic leptin resistance and long-term energy homeostasis. Diabetes. 2010;59:894-906; PMID:20068134. doi:10.2337/db09-1024.
  • Howard JK, Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins. Trends Endocrinol Metab. 2006;17:365-371; PMID:17010638. doi:10.1016/j.tem.2006.09.007.
  • Augustine RA, Grattan DR. Induction of central leptin resistance in hyperphagic pseudopregnant rats by chronic prolactin infusion. Endocrinology. 2008;149:1049-1055; PMID:18063686. doi:10.1210/en.2007-1018.
  • Naef L, Woodside B. Prolactin/leptin interactions in the control of food intake in rats. Endocrinology. 2007;148:5977-5983; PMID:17872372. doi:10.1210/en.2007-0442.
  • Anderson GM, Beijer P, Bang AS, Fenwick MA, Bunn SJ, Grattan DR. Suppression of prolactin-induced signal transducer and activator of transcription 5b signaling and induction of suppressors of cytokine signaling messenger ribonucleic acid in the hypothalamic arcuate nucleus of the rat during late pregnancy and lactation. Endocrinology. 2006;147:4996-5005; PMID:16857756. doi:10.1210/en.2005-0755.
  • St-Pierre J, Tremblay ML. Modulation of leptin resistance by protein tyrosine phosphatases. Cell Metab. 2012;15:292-297; PMID:22405067. doi:10.1016/j.cmet.2012.02.004.
  • Bence KK, Delibegovic M, Xue B, Gorgun CZ, Hotamisligil GS, Neel BG, Kahn BB. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med. 2006;12:917-924; PMID:16845389. doi:10.1038/nm1435.
  • Kaszubska W, Falls HD, Schaefer VG, Haasch D, Frost L, Hessler P, Kroeger PE, White DW, Jirousek MR, Trevillyan JM. Protein tyrosine phosphatase 1B negatively regulates leptin signaling in a hypothalamic cell line. Mol Cell Endocrinol. 2002;195:109-118; PMID:12354677. doi:10.1016/S0303-7207(02)00178-8.
  • Loh K, Fukushima A, Zhang X, Galic S, Briggs D, Enriori PJ, Simonds S, Wiede F, Reichenbach A, Hauser C, et al. Elevated hypothalamic TCPTP in obesity contributes to cellular leptin resistance. Cell Metab. 2011;14:684-699; PMID:22000926. doi:10.1016/j.cmet.2011.09.011.
  • Rousso-Noori L, Knobler H, Levy-Apter E, Kuperman Y, Neufeld-Cohen A, Keshet Y, Akepati VR, Klinghoffer RA, Chen A, Elson A. Protein tyrosine phosphatase epsilon affects body weight by downregulating leptin signaling in a phosphorylation-dependent manner. Cell Metab. 2011;13:562-572; PMID:21531338. doi:10.1016/j.cmet.2011.02.017.
  • Tsou RC, Zimmer DJ, De Jonghe BC, Bence KK. Deficiency of PTP1B in leptin receptor-expressing neurons leads to decreased body weight and adiposity in mice. Endocrinology. 2012;153:4227-4237; PMID:22802463. doi:10.1210/en.2012-1548.
  • Zhang EE, Chapeau E, Hagihara K, Feng GS. Neuronal Shp2 tyrosine phosphatase controls energy balance and metabolism. Proc Natl Acad Sci USA. 2004;101:16064-16069; PMID:15520383. doi:10.1073/pnas.0405041101.
  • Banno R, Zimmer D, De Jonghe BC, Atienza M, Rak K, Yang W, Bence KK. PTP1B and SHP2 in POMC neurons reciprocally regulate energy balance in mice. J Clin Invest. 2010;120:720-734; PMID:20160350. doi:10.1172/JCI39620.
  • Morris DL, Rui L. Recent advances in understanding leptin signaling and leptin resistance. Am J Physiol Endocrinol Metab. 2009;297:E1247-E1259; PMID:19724019. doi:10.1152/ajpendo.00274.2009.
  • Ren D, Zhou Y, Morris D, Li M, Li Z, Rui L. Neuronal SH2B1 is essential for controlling energy and glucose homeostasis. J Clin Invest. 2007;117:397-406; PMID:17235396. doi:10.1172/JCI29417.
  • Velloso LA, Folli F, Saad MJ. TLR4 at the crossroads of nutrients, gut microbiota, and metabolic inflammation. Endocr Rev. 2015;36:245-271; PMID:25811237. doi:10.1210/er.2014-1100.
  • Cnop M, Foufelle F, Velloso LA. Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med. 2012;18:59-68; PMID:21889406. doi:10.1016/j.molmed.2011.07.010.
  • De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC, Saad MJ, Velloso LA. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology. 2005;146:4192-4199; PMID:16002529. doi:10.1210/en.2004-1520.
  • Moraes JC, Coope A, Morari J, Cintra DE, Roman EA, Pauli JR, Romanatto T, Carvalheira JB, Oliveira AL, Saad MJ, et al. High-fat diet induces apoptosis of hypothalamic neurons. PLoS One. 2009;4:e5045; PMID:19340313. doi:10.1371/journal.pone.0005045.
  • McNay DEG, Briancon N, Kokoeva MV, Maratos-Flier E, Flier JS. Remodeling of the arcuate nucleus energy-balance circuit is inhibited in obese mice. J Clin Invest. 2012;122:142-152; PMID:22201680. doi:10.1172/JCI43134.
  • Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, Tsukumo DM, Anhe G, Amaral ME, Takahashi HK, et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. J Neurosci. 2009;29:359-370; PMID:19144836. doi:10.1523/JNEUROSCI.2760-08.2009.
  • Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell. 2008;135:61-73; PMID:18854155. doi:10.1016/j.cell.2008.07.043.
  • Amaral ME, Barbuio R, Milanski M, Romanatto T, Barbosa HC, Nadruz W, Bertolo MB, Boschero AC, Saad MJ, Franchini KG, et al. Tumor necrosis factor-alpha activates signal transduction in hypothalamus and modulates the expression of pro-inflammatory proteins and orexigenic/anorexigenic neurotransmitters. J Neurochem. 2006;98:203-212; PMID:16638016. doi:10.1111/j.1471-4159.2006.03857.x.
  • Nakamura T, Furuhashi M, Li P, Cao H, Tuncman G, Sonenberg N, Gorgun CZ, Hotamisligil GS. Double-stranded RNA-dependent protein kinase links pathogen sensing with stress and metabolic homeostasis. Cell. 2010;140:338-348; PMID:20144759. doi:10.1016/j.cell.2010.01.001.
  • Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, Myers MG Jr, Ozcan U. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9:35-51; PMID:19117545. doi:10.1016/j.cmet.2008.12.004.
  • Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii M, Ozawa K. Endoplasmic reticulum stress induces leptin resistance. Mol Pharmacol. 2008;74:1610-1619; PMID:18755873. doi:10.1124/mol.108.050070.
  • de Git KC, Adan RA. Leptin resistance in diet-induced obesity: the role of hypothalamic inflammation. Obes Rev. 2015;16:207-224; PMID:25589226. doi:10.1111/obr.12243.
  • Balland E, Cowley MA. New insights in leptin resistance mechanisms in mice. Front Neuroendocrinol. 2015;39:59-65; PMID:26410445. doi:10.1016/j.yfrne.2015.09.004.
  • Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027-1031; PMID:17183312. doi:10.1038/nature05414.
  • Claesson MJ, Jeffery IB, Conde S, Power SE, O'Connor EM, Cusack S, Harris HM, Coakley M, Lakshminarayanan B, O'Sullivan O, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178-184; PMID:22797518. doi:10.1038/nature11319.
  • Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500:585-588; PMID:23985875. doi:10.1038/nature12480.
  • Johnson AM, Olefsky JM. The origins and drivers of insulin resistance. Cell. 2013;152:673-684; PMID:23415219. doi:10.1016/j.cell.2013.01.041.
  • Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, Israeli D, Zmora N, Gilad S, Weinberger A, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514:181-186; PMID:25231862. doi:10.1038/nature13793.
  • Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, Gewirtz AT. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature. 2015;519:92-96; PMID:25731162. doi:10.1038/nature14232.
  • Donato J Jr The central nervous system as a promising target to treat diabetes mellitus. Curr Top Med Chem. 2012;12:2070-2081; PMID:23167796. doi:10.2174/156802612804910214.
  • Hung HY, Qian K, Morris-Natschke SL, Hsu CS, Lee KH. Recent discovery of plant-derived anti-diabetic natural products. Nat Prod Rep. 2012;29:580-606; PMID:22491825. doi:10.1039/c2np00074a.
  • Sasaki T. Age-associated weight gain, Leptin, and SIRT1. A possible role for hypothalamic SIRT1 in the prevention of weight gain and aging through modulation of leptin sensitivity. Front Endocrinol (Lausanne). 2015;6:109; PMID:26236282. doi:10.3389/fendo.2015.00109.
  • Lee J, Liu J, Feng X, Salazar Hernandez MA, Mucka P, Ibi D, Choi JW, Ozcan U. Withaferin A is a leptin sensitizer with strong antidiabetic properties in mice. Nat Med. 2016;22:1023-1032; PMID:27479085. doi:10.1038/nm.4145.
  • Liu J, Lee J, Salazar Hernandez MA, Mazitschek R, Ozcan U. Treatment of obesity with celastrol. Cell. 2015;161:999-1011; PMID:26000480. doi:10.1016/j.cell.2015.05.011.
  • Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, Anderson CM, Parkes DG, Baron AD. Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci USA. 2008;105:7257-7262; PMID:18458326. doi:10.1073/pnas.0706473105.
  • Trevaskis JL, Coffey T, Cole R, Lei C, Wittmer C, Walsh B, Weyer C, Koda J, Baron AD, Parkes DG, et al. Amylin-mediated restoration of leptin responsiveness in diet-induced obesity: magnitude and mechanisms. Endocrinology. 2008;149:5679-5687; PMID:18669592. doi:10.1210/en.2008-0770.
  • Trevaskis JL, Lei C, Koda JE, Weyer C, Parkes DG, Roth JD. Interaction of leptin and amylin in the long-term maintenance of weight loss in diet-induced obese rats. Obesity. 2009;18:21-26; PMID:19543217. doi:10.1038/oby.2009.187.
  • Kusakabe T, Ebihara K, Sakai T, Miyamoto L, Aotani D, Yamamoto Y, Yamamoto-Kataoka S, Aizawa-Abe M, Fujikura J, Hosoda K, et al. Amylin improves the effect of leptin on insulin sensitivity in leptin-resistant diet-induced obese mice. Am J Physiol Endocrinol Metab. 2012;302:E924-E931; PMID:22275759. doi:10.1152/ajpendo.00198.2011.
  • Muller TD, Sullivan LM, Habegger K, Yi CX, Kabra D, Grant E, Ottaway N, Krishna R, Holland J, Hembree J, et al. Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in combination with exendin-4 or FGF21. J Pept Sci. 2012;18:383-393; PMID:22565812. doi:10.1002/psc.2408.
  • Cintra DE, Ropelle ER, Moraes JC, Pauli JR, Morari J, de Souza CT, Grimaldi R, Stahl M, Carvalheira JB, Saad MJ, et al. Unsaturated fatty acids revert diet-induced hypothalamic inflammation in obesity. PLoS One. 2012;7:e30571; PMID:22279596. doi:10.1371/journal.pone.0030571.
  • Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 2010;142:687-698; PMID:20813258. doi:10.1016/j.cell.2010.07.041.
  • Ichimura A, Hirasawa A, Poulain-Godefroy O, Bonnefond A, Hara T, Yengo L, Kimura I, Leloire A, Liu N, Iida K, et al. Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human. Nature. 2012;483:350-354; PMID:22343897. doi:10.1038/nature10798.
  • Nascimento LF, Souza GF, Morari J, Barbosa GO, Solon C, Moura RF, Victório SC, Ignácio-Souza LM, Razolli DS, Carvalho HF, et al. n-3 fatty acids induce neurogenesis of predominantly POMC-Expressing cells in the hypothalamus. Diabetes. 2016;65:673-686; PMID:26512023. doi:10.2337/db15-0008.
  • Steinberg GR, Smith AC, Wormald S, Malenfant P, Collier C, Dyck DJ. Endurance training partially reverses dietary-induced leptin resistance in rodent skeletal muscle. Am J Physiol Endocrinol Metab. 2004;286:E57-E63; PMID:14662513. doi:10.1152/ajpendo.00302.2003.
  • Kang S, Kim KB, Shin KO. Exercise training improves leptin sensitivity in peripheral tissue of obese rats. Biochem Biophys Res Commun. 2013;435:454-459; PMID:23669042. doi:10.1016/j.bbrc.2013.05.007.
  • Carhuatanta KAK, Demuro G, Tschöp MH, Pfluger PT, Benoit SC, Obici S. Voluntary exercise improves high-fat diet-induced leptin resistance independent of adiposity. Endocrinology. 2011;152:2655-2664; PMID:21586558. doi:10.1210/en.2010-1340.
  • Laing BT, Do K, Matsubara T, Wert DW, Avery MJ, Langdon EM, Zheng D, Huang H. Voluntary exercise improves hypothalamic and metabolic function in obese mice. J Endocrinol. 2016;229:109-122; PMID:26931136. doi:10.1530/JOE-15-0510.
  • Patterson CM, Bouret SG, Dunn-Meynell AA, Levin BE. Three weeks of postweaning exercise in DIO rats produces prolonged increases in central leptin sensitivity and signaling. Am J Physiol Regul Integr Comp Physiol. 2009;296:R537-R548; PMID:19158409. doi:10.1152/ajpregu.90859.2008.
  • Chiarreotto-Ropelle EC, Pauli LS, Katashima CK, Pimentel GD, Picardi PK, Silva VR, de Souza CT, Prada PO, Cintra DE, Carvalheira JB, et al. Acute exercise suppresses hypothalamic PTP1B protein level and improves insulin and leptin signaling in obese rats. Am J Physiol Endocrinol Metab. 2013;305:E649-E659; PMID:23880311. doi:10.1152/ajpendo.00272.2013.
  • McMillen IC, Robinson JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005;85:571-633; PMID:15788706. doi:10.1152/physrev.00053.2003.
  • Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull. 2001;60:5-20; PMID:11809615. doi:10.1093/bmb/60.1.5.
  • Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine exposure in utero and early infancy. N Engl J Med. 1976;295:349-353; PMID:934222. doi:10.1056/NEJM197608122950701.
  • Baquero AF, de Solis AJ, Lindsley SR, Kirigiti MA, Smith MS, Cowley MA, Zeltser LM, Grove KL. Developmental switch of leptin signaling in arcuate nucleus neurons. J Neurosci. 2014;34:9982-9994; PMID:25057200. doi:10.1523/JNEUROSCI.0933-14.2014.
  • Juan De Solis A, Baquero AF, Bennett CM, Grove KL, Zeltser LM. Postnatal undernutrition delays a key step in the maturation of hypothalamic feeding circuits. Mol Metab. 2016;5:198-209; PMID:26977392. doi:10.1016/j.molmet.2016.01.003.
  • Ralevski A, Horvath TL. Developmental programming of hypothalamic neuroendocrine systems. Front Neuroendocrinol. 2015;39:52-58; PMID:26391503. doi:10.1016/j.yfrne.2015.09.002.
  • Guo F, Jen KL. High-fat feeding during pregnancy and lactation affects offspring metabolism in rats. Physiol Behav. 1995;57:681-686; PMID:7777603. doi:10.1016/0031-9384(94)00342-4.
  • Bayol SA, Farrington SJ, Stickland NC. A maternal ‘junk food’ diet in pregnancy and lactation promotes an exacerbated taste for ‘junk food’ and a greater propensity for obesity in rat offspring. Br J Nutr. 2007;98:843-851; PMID:17697422. doi:10.1017/S0007114507812037.
  • Vogt Merly C, Paeger L, Hess S, Steculorum Sophie M, Awazawa M, Hampel B, Neupert S, Nicholls HT, Mauer J, Hausen AC, et al. Neonatal insulin action impairs hypothalamic neurocircuit formation in response to maternal high-fat feeding. Cell. 2014;156:495-509; PMID:24462248. doi:10.1016/j.cell.2014.01.008.
  • Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290-e296; PMID:15741354. doi:10.1542/peds.2004-1808.
  • Attig L, Solomon G, Ferezou J, Abdennebi-Najar L, Taouis M, Gertler A, Djiane J. Early postnatal leptin blockage leads to a long-term leptin resistance and susceptibility to diet-induced obesity in rats. Int J Obes (Lond). 2008;32:1153-1160; PMID:18379577. doi:10.1038/ijo.2008.39.
  • Jaquet D, Leger J, Levy-Marchal C, Oury JF, Czernichow P. Ontogeny of leptin in human fetuses and newborns: effect of intrauterine growth retardation on serum leptin concentrations. J Clin Endocrinol Metab. 1998;83:1243-1246; PMID:9543149. doi:10.1210/jcem.83.4.4731.
  • Vickers MH, Gluckman PD, Coveny AH, Hofman PL, Cutfield WS, Gertler A, Breier BH, Harris M. Neonatal leptin treatment reverses developmental programming. Endocrinology. 2005;146:4211-4216; PMID:16020474. doi:10.1210/en.2005-0581.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.