Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front Med    2013, Vol. 7 Issue (2) : 207-222    https://doi.org/10.1007/s11684-013-0263-5
REVIEW
Leptin signaling and leptin resistance
Yingjiang Zhou1(), Liangyou Rui2()
1. Cambridge Laboratories, Pfizer Worldwide Research & Development, Cambridge, MA 02139, USA; 2. Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
 Download: PDF(405 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Leptin is secreted into the bloodstream by adipocytes and is required for the maintenance of energy homeostasis and body weight. Leptin deficiency or genetic defects in the components of the leptin signaling pathways cause obesity. Leptin controls energy balance and body weight mainly through leptin receptor b (LEPRb)-expressing neurons in the brain, particularly in the hypothalamus. These LEPRb-expressing neurons function as the first-order neurons that project to the second-order neurons located within and outside the hypothalamus, forming a neural network that controls the energy homeostasis and body weight. Multiple factors, including inflammation and endoplasmic reticulum (ER) stress, contribute to leptin resistance. Leptin resistance is the key risk factor for obesity. This review is focused on recent advance about leptin action, leptin signaling, and leptin resistance.

Keywords leptin signaling      leptin receptor      energy balance      leptin resistance      obesity     
Corresponding Author(s): Zhou Yingjiang,Email:yingjiang.zhou@pfizer.com; Rui Liangyou,Email:ruily@umich.edu   
Issue Date: 05 June 2013
 Cite this article:   
Yingjiang Zhou,Liangyou Rui. Leptin signaling and leptin resistance[J]. Front Med, 2013, 7(2): 207-222.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-013-0263-5
https://academic.hep.com.cn/fmd/EN/Y2013/V7/I2/207
Fig.1  Leptin signaling pathways. Leptin binds to LEPRb and activates JAK2. JAK2 phosphorylates LEPRb on Tyr, Tyr and Tyr. Phospho-Tyr, -Tyr and-Tyr bind to downstream molecules and activate the JAK2/STAT3, JAK2/STAT5, PI3K/IRS/AKT, and SHP2/ERK pathways. These pathways act coordinately to regulate energy balance and body weight. LEPRb signaling is regulated both negatively by SOCS3, PTP1B, TCPTP and PTEN and positively by SH2B1. Many factors, including hyperleptinemia, inflammation, endoplasmic reticulum (ER) stress, and defective autophagy, contribute to leptin resistance.
Fig.2  Leptin-targeted neurons and neural circuits. Leptin directly suppresses NPY/AgRP neurons and stimulates POMC/CART neurons in the ARC. ARC neurons project to multiple hypothalamic areas including the DMH, VMH, PVH, and LHA. Leptin also directly activates LEPRb in DMH, VMH, PVH, and LHA neurons. The PVH and LHA are important hypothalamic output pathways that mediate leptin’s anti-obesity action. VTA: ventral tegmental area; NTS: solitary nucleus; IML: intermediolateral cell column.
1 Kopelman PG. Obesity as a medical problem. Nature 2000; 404(6778): 635–643
pmid:10766250
2 Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, Singh GM, Gutierrez HR, Lu Y, Bahalim AN, Farzadfar F, Riley LM, Ezzati M; the Global Burden of Metabolic Risk Factors of Chronic Diseases Collaborating Group (Body Mass Index) . National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9·1 million participants. Lancet 2011; 377(9765): 557–567
doi: 10.1016/S0140-6736(10)62037-5 pmid:21295846
3 Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature 2000; 404(6778): 661–671
pmid:10766253
4 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(6505): 425–432
doi: 10.1038/372425a0 pmid:7984236
5 Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI. Identification and expression cloning of a leptin receptor, OB-R. Cell 1995; 83(7): 1263–1271
doi: 10.1016/0092-8674(95)90151-5 pmid:8548812
6 Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O’Rahilly S. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 1997; 387(6636): 903–908
doi: 10.1038/43185 pmid:9202122
7 Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin missense mutation associated with hypogonadism and morbid obesity. Nat Genet 1998; 18(3): 213–215
doi: 10.1038/ng0398-213 pmid:9500540
8 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(12): 879–884
doi: 10.1056/NEJM199909163411204 pmid:10486419
9 Gibson WT, Farooqi IS, Moreau M, DePaoli AM, Lawrence E, O’Rahilly S, Trussell RA. Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J Clin Endocrinol Metab 2004; 89(10): 4821–4826
doi: 10.1210/jc.2004-0376 pmid:15472169
10 Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, O’Rahilly S. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 2002; 110(8): 1093–1103
pmid:12393845
11 Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O’Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, Krauss RM, Veldhuis JD, Wagner AJ, DePaoli AM, McCann SM, Wong ML. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA 2004; 101(13): 4531–4536
doi: 10.1073/pnas.0308767101 pmid:15070752
12 Zhang F, Basinski MB, Beals JM, Briggs SL, Churgay LM, Clawson DK, DiMarchi RD, Furman TC, Hale JE, Hsiung HM, Schoner BE, Smith DP, Zhang XY, Wery JP, Schevitz RW. Crystal structure of the obese protein leptin-E100. Nature 1997; 387(6629): 206–209
doi: 10.1038/387206a0 pmid:9144295
13 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(11): 1407–1433
doi: 10.1038/sj.ijo.0802142 pmid:12439643
14 Wrann CD, Eguchi J, Bozec A, Xu Z, Mikkelsen T, Gimble J, Nave H, Wagner EF, Ong SE, Rosen ED. FOSL2 promotes leptin gene expression in human and mouse adipocytes. J Clin Invest 2012; 122(3): 1010–1021
doi: 10.1172/JCI58431 pmid:22326952
15 Frederich RC, Hamann A, Anderson S, L?llmann B, Lowell BB, Flier JS. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat Med 1995; 1(12): 1311–1314
doi: 10.1038/nm1295-1311 pmid:7489415
16 Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, Kim S, Lallone R, Ranganathan S, Kern PA, Friedman JM. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med 1995; 1(11): 1155–1161
doi: 10.1038/nm1195-1155 pmid:7584987
17 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(6588): 250–252
doi: 10.1038/382250a0 pmid:8717038
18 Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim SY, Hamnvik OP, Koniaris A. Leptin in human physiology and pathophysiology. Am J Physiol Endocrinol Metab 2011; 301(4): E567–E584
doi: 10.1152/ajpendo.00315.2011 pmid:21791620
19 Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998; 395(6704): 763–770
doi: 10.1038/27376 pmid:9796811
20 Bates SH, Myers MG Jr. The role of leptin receptor signaling in feeding and neuroendocrine function. Trends Endocrinol Metab 2003; 14(10): 447–452
doi: 10.1016/j.tem.2003.10.003 pmid:14643059
21 Gautron L, Elmquist JK. Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 2011; 121(6): 2087–2093
doi: 10.1172/JCI45888 pmid:21633176
22 Elmquist JK, Bj?rbaek C, Ahima RS, Flier JS, Saper CB. Distributions of leptin receptor mRNA isoforms in the rat brain. J Comp Neurol 1998; 395(4): 535–547
doi: 10.1002/(SICI)1096-9861(19980615)395:4<535::AID-CNE9>3.0.CO;2-2 pmid:9619505
23 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(5): 518–532
doi: 10.1002/cne.22025 pmid:19350671
24 Schwartz MW, Seeley RJ, Campfield LA, Burn P, Baskin DG. Identification of targets of leptin action in rat hypothalamus. J Clin Invest 1996; 98(5): 1101–1106
doi: 10.1172/JCI118891 pmid:8787671
25 Shpilman M, Niv-Spector L, Katz M, Varol C, Solomon G, Ayalon-Soffer M, Boder E, Halpern Z, Elinav E, Gertler A. Development and characterization of high affinity leptins and leptin antagonists. J Biol Chem 2011; 286(6): 4429–4442
doi: 10.1074/jbc.M110.196402 pmid:21119198
26 Elinav E, Niv-Spector L, Katz M, Price TO, Ali M, Yacobovitz M, Solomon G, Reicher S, Lynch JL, Halpern Z, Banks WA, Gertler A. Pegylated leptin antagonist is a potent orexigenic agent: preparation and mechanism of activity. Endocrinology 2009; 150(7): 3083–3091
doi: 10.1210/en.2008-1706 pmid:19342450
27 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(13): 7001–7005
doi: 10.1073/pnas.94.13.7001 pmid:9192681
28 Morris DL, Rui L. Recent advances in understanding leptin signaling and leptin resistance. Am J Physiol Endocrinol Metab 2009; 297(6): E1247–E1259
doi: 10.1152/ajpendo.00274.2009 pmid:19724019
29 Myers MG, Cowley MA, Münzberg H. Mechanisms of leptin action and leptin resistance. Annu Rev Physiol 2008; 70(1): 537–556
doi: 10.1146/annurev.physiol.70.113006.100707 pmid:17937601
30 Baumann H, Morella KK, White DW, Dembski M, Bailon PS, Kim H, Lai CF, Tartaglia LA. The full-length leptin receptor has signaling capabilities of interleukin 6-type cytokine receptors. Proc Natl Acad Sci USA 1996; 93(16): 8374–8378
doi: 10.1073/pnas.93.16.8374 pmid:8710878
31 Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, Duyk GM, Tepper RI, Morgenstern JP. 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(3): 491–495
doi: 10.1016/S0092-8674(00)81294-5 pmid:8608603
32 Chua SC Jr, Chung WK, Wu-Peng XS, Zhang Y, Liu SM, Tartaglia L, Leibel RL. Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 1996; 271(5251): 994–996
doi: 10.1126/science.271.5251.994 pmid:8584938
33 Uotani S, Bj?rbaek C, Torn?e J, Flier JS. Functional properties of leptin receptor isoforms: internalization and degradation of leptin and ligand-induced receptor downregulation. Diabetes 1999; 48(2): 279–286
doi: 10.2337/diabetes.48.2.279 pmid:10334302
34 Schaab M, Kausch H, Klammt J, Nowicki M, Anderegg U, Gebhardt R, Rose-John S, Scheller J, Thiery J, Kratzsch J. Novel regulatory mechanisms for generation of the soluble leptin receptor: implications for leptin action. PLoS ONE 2012; 7(4): e34787
doi: 10.1371/journal.pone.0034787 pmid:22545089
35 Taga T, Kishimoto T. Gp130 and the interleukin-6 family of cytokines. Annu Rev Immunol 1997; 15(1): 797–819
doi: 10.1146/annurev.immunol.15.1.797 pmid:9143707
36 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(19): 14563–14572
doi: 10.1074/jbc.275.19.14563 pmid:10799542
37 Tartaglia LA. The leptin receptor. J Biol Chem 1997; 272(10): 6093–6096
pmid:9102398
38 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(1): 109–119
doi: 10.1111/j.1432-1033.2004.04391.x pmid:15634336
39 White DW, Kuropatwinski KK, Devos R, Baumann H, Tartaglia LA. Leptin receptor (OB-R) signaling. Cytoplasmic domain mutational analysis and evidence for receptor homo-oligomerization. J Biol Chem 1997; 272(7): 4065–4071
doi: 10.1074/jbc.272.7.4065 pmid:9020115
40 Jiang L, Li Z, Rui L. Leptin stimulates both JAK2-dependent and JAK2-independent signaling pathways. J Biol Chem 2008; 283(42): 28066–28073
doi: 10.1074/jbc.M805545200 pmid:18718905
41 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(1): 95–97
doi: 10.1038/ng0996-95 pmid:8782827
42 Xu AW, Ste-Marie L, Kaelin CB, Barsh GS. Inactivation of signal transducer and activator of transcription 3 in proopiomelanocortin (Pomc) neurons causes decreased pomc expression, mild obesity, and defects in compensatory refeeding. Endocrinology 2007; 148(1): 72–80
doi: 10.1210/en.2006-1119 pmid:17023536
43 Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, Neel BG, Schwartz MW, Myers MG Jr. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 2003; 421(6925): 856–859
doi: 10.1038/nature01388 pmid:12594516
44 Jiang L, You J, Yu X, Gonzalez L, Yu Y, Wang Q, Yang G, Li W, Li C, Liu Y. Tyrosine-dependent and-independent actions of leptin receptor in control of energy balance and glucose homeostasis. Proc Natl Acad Sci USA 2008; 105(47): 18619–18624
doi: 10.1073/pnas.0804589105 pmid:19015522
45 Piper ML, Unger EK, Myers MG Jr, Xu AW. Specific physiological roles for signal transducer and activator of transcription 3 in leptin receptor-expressing neurons. Mol Endocrinol 2008; 22(3): 751–759
doi: 10.1210/me.2007-0389 pmid:18096691
46 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(13): 4661–4666
doi: 10.1073/pnas.0303992101 pmid:15070774
47 Gong Y, Ishida-Takahashi R, Villanueva EC, Fingar DC, Münzberg H, Myers MG Jr. The long form of the leptin receptor regulates STAT5 and ribosomal protein S6 via alternate mechanisms. J Biol Chem 2007; 282(42): 31019–31027
doi: 10.1074/jbc.M702838200 pmid:17726024
48 Mütze J, Roth J, Gerstberger R, Hübschle T. Nuclear translocation of the transcription factor STAT5 in the rat brain after systemic leptin administration. Neurosci Lett 2007; 417(3): 286–291
doi: 10.1016/j.neulet.2007.02.074 pmid:17353091
49 Lee J Y, Muenzberg H, Gavrilova O, Reed J A, Berryman D, Villanueva E C, Louis G W, Leinninger G M, Bertuzzi S, Seeley R J, Robinson G W, Myers M G, Hennighausen L.Loss of cytokine-STAT5 signaling in the CNS and pituitary gland alters energy balance and leads to obesity. PLoS One 2008; 3(2): e1639
doi: 10.1371/journal.pone.0001639 pmid:18286195
50 Bj?rbaek C, Buchholz RM, Davis SM, Bates SH, Pierroz DD, Gu H, Neel BG, Myers MG Jr, Flier JS. Divergent roles of SHP-2 in ERK activation by leptin receptors. J Biol Chem 2001; 276(7): 4747–4755
doi: 10.1074/jbc.M007439200 pmid:11085989
51 Rahmouni K, Sigmund CD, Haynes WG, Mark AL. Hypothalamic ERK mediates the anorectic and thermogenic sympathetic effects of leptin. Diabetes 2009; 58(3): 536–542
doi: 10.2337/db08-0822 pmid:19066310
52 Li C, Friedman JM. Leptin receptor activation of SH2 domain containing protein tyrosine phosphatase 2 modulates Ob receptor signal transduction. Proc Natl Acad Sci USA 1999; 96(17): 9677–9682
doi: 10.1073/pnas.96.17.9677 pmid:10449753
53 He Z, Zhang SS, Meng Q, Li S, Zhu HH, Raquil MA, Alderson N, Zhang H, Wu J, Rui L, Cai D, Feng GS. Shp2 controls female body weight and energy balance by integrating leptin and estrogen signals. Mol Cell Biol 2012; 32(10): 1867–1878
doi: 10.1128/MCB.06712-11 pmid:22431513
54 Krajewska M, Banares S, Zhang EE, Huang X, Scadeng M, Jhala US, Feng GS, Krajewski S. Development of diabesity in mice with neuronal deletion of Shp2 tyrosine phosphatase. Am J Pathol 2008; 172(5): 1312–1324
doi: 10.2353/ajpath.2008.070594 pmid:18403587
55 St-Pierre J, Tremblay ML. Modulation of leptin resistance by protein tyrosine phosphatases. Cell Metab 2012; 15(3): 292–297
doi: 10.1016/j.cmet.2012.02.004 pmid:22405067
56 Zhang EE, Chapeau E, Hagihara K, Feng GS. Neuronal Shp2 tyrosine phosphatase controls energy balance and metabolism. Proc Natl Acad Sci USA 2004; 101(45): 16064–16069
doi: 10.1073/pnas.0405041101 pmid:15520383
57 Bjorbak C, Lavery HJ, Bates SH, Olson RK, Davis SM, Flier JS, Myers MG Jr. SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem 2000; 275(51): 40649–40657
doi: 10.1074/jbc.M007577200 pmid:11018044
58 You J, Yu Y, Jiang L, Li W, Yu X, Gonzalez L, Yang G, Ke Z, Li W, Li C, Liu Y. Signaling through Tyr985 of leptin receptor as an age/diet-dependent switch in the regulation of energy balance. Mol Cell Biol 2010; 30(7): 1650–1659
doi: 10.1128/MCB.01307-09 pmid:20086094
59 Bj?rnholm M, Münzberg H, Leshan RL, Villanueva EC, Bates SH, Louis GW, Jones JC, Ishida-Takahashi R, Bj?rbaek C, Myers MG Jr. Mice lacking inhibitory leptin receptor signals are lean with normal endocrine function. J Clin Invest 2007; 117(5): 1354–1360
doi: 10.1172/JCI30688 pmid:17415414
60 Niswender KD, Morton GJ, Stearns WH, Rhodes CJ, Myers MG Jr, Schwartz MW. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature 2001; 413(6858): 794–795
doi: 10.1038/35101657 pmid:11677594
61 Kim YB, Uotani S, Pierroz DD, Flier JS, Kahn BB. In vivo administration of leptin activates signal transduction directly in insulin-sensitive tissues: overlapping but distinct pathways from insulin. Endocrinology 2000; 141(7): 2328–2339
doi: 10.1210/en.141.7.2328 pmid:10875232
62 Zhao AZ, Huan JN, Gupta S, Pal R, Sahu A. A phosphatidylinositol 3-kinase phosphodiesterase 3B-cyclic AMP pathway in hypothalamic action of leptin on feeding. Nat Neurosci 2002; 5(8): 727–728
pmid:12101402
63 Xu AW, Kaelin CB, Takeda K, Akira S, Schwartz MW, Barsh GS. PI3K integrates the action of insulin and leptin on hypothalamic neurons. J Clin Invest 2005; 115(4): 951–958
pmid:15761497
64 Lin X, Taguchi A, Park S, Kushner JA, Li F, Li Y, White MF. Dysregulation of insulin receptor substrate 2 in β cells and brain causes obesity and diabetes. J Clin Invest 2004; 114(7): 908–916
pmid:15467829
65 Kubota N, Terauchi Y, Tobe K, Yano W, Suzuki R, Ueki K, Takamoto I, Satoh H, Maki T, Kubota T, Moroi M, Okada-Iwabu M, Ezaki O, Nagai R, Ueta Y, Kadowaki T, Noda T. Insulin receptor substrate 2 plays a crucial role in βcells and the hypothalamus. J Clin Invest 2004; 114(7): 917–927
pmid:15467830
66 Duan C, Li M, Rui L. SH2-B promotes insulin receptor substrate 1 (IRS1)- and IRS2-mediated activation of the phosphatidylinositol 3-kinase pathway in response to leptin. J Biol Chem 2004; 279(42): 43684–43691
doi: 10.1074/jbc.M408495200 pmid:15316008
67 Ren D, Li M, Duan C, Rui L. Identification of SH2-B as a key regulator of leptin sensitivity, energy balance, and body weight in mice. Cell Metab 2005; 2(2): 95–104
doi: 10.1016/j.cmet.2005.07.004 pmid:16098827
68 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(2): 397–406
doi: 10.1172/JCI29417 pmid:17235396
69 Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 2006; 7(2): 85–96
doi: 10.1038/nrm1837 pmid:16493415
70 Kim MS, Pak YK, Jang PG, Namkoong C, Choi YS, Won JC, Kim KS, Kim SW, Kim HS, Park JY, Kim YB, Lee KU. Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis. Nat Neurosci 2006; 9(7): 901–906
doi: 10.1038/nn1731 pmid:16783365
71 Kitamura T, Feng Y, Kitamura YI, Chua SC Jr, Xu AW, Barsh GS, Rossetti L, Accili D. Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat Med 2006; 12(5): 534–540
doi: 10.1038/nm1392 pmid:16604086
72 Plum L, Lin HV, Dutia R, Tanaka J, Aizawa KS, Matsumoto M, Kim AJ, Cawley NX, Paik JH, Loh YP, DePinho RA, Wardlaw SL, Accili D. The obesity susceptibility gene Cpe links FoxO1 signaling in hypothalamic pro-opiomelanocortin neurons with regulation of food intake. Nat Med 2009; 15(10): 1195–1201
doi: 10.1038/nm.2026 pmid:19767734
73 Sadagurski M, Leshan RL, Patterson C, Rozzo A, Kuznetsova A, Skorupski J, Jones JC, Depinho RA, Myers MG Jr, White MF. IRS2 signaling in LepR-b neurons suppresses FoxO1 to control energy balance independently of leptin action. Cell Metab 2012; 15(5): 703–712
doi: 10.1016/j.cmet.2012.04.011 pmid:22560222
74 Yang G, Lim CY, Li C, Xiao X, Radda GK, Li C, Cao X, Han W. FoxO1 inhibits leptin regulation of pro-opiomelanocortin promoter activity by blocking STAT3 interaction with specificity protein 1. J Biol Chem 2009; 284(6): 3719–3727
doi: 10.1074/jbc.M804965200 pmid:19049975
75 Cota D, Proulx K, Smith KA, Kozma SC, Thomas G, Woods SC, Seeley RJ. Hypothalamic mTOR signaling regulates food intake. Science 2006; 312(5775): 927–930
doi: 10.1126/science.1124147 pmid:16690869
76 Maya-Monteiro CM, Bozza PT. Leptin and mTOR: partners in metabolism and inflammation. Cell Cycle 2008; 7(12): 1713–1717
doi: 10.4161/cc.7.12.6157 pmid:18583936
77 Blouet C, Ono H, Schwartz GJ. Mediobasal hypothalamic p70 S6 kinase 1 modulates the control of energy homeostasis. Cell Metab 2008; 8(6): 459–467
doi: 10.1016/j.cmet.2008.10.004 pmid:19041762
78 Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, Mu J, Foufelle F, Ferré P, Birnbaum MJ, Stuck BJ, Kahn BB. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 2004; 428(6982): 569–574
doi: 10.1038/nature02440 pmid:15058305
79 Dagon Y, Hur E, Zheng B, Wellenstein K, Cantley LC, Kahn BB. p70S6 kinase phosphorylates AMPK on serine 491 to mediate leptin’s effect on food intake. Cell Metab 2012; 16(1): 104–112
doi: 10.1016/j.cmet.2012.05.010 pmid:22727014
80 Su H, Jiang L, Carter-Su C, Rui L. Glucose enhances leptin signaling through modulation of AMPK activity. PLoS ONE 2012; 7(2): e31636
doi: 10.1371/journal.pone.0031636 pmid:22359610
81 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(16): 5240–5250
doi: 10.1523/JNEUROSCI.0405-09.2009 pmid:19386920
82 Leinninger GM, Jo YH, Leshan RL, Louis GW, Yang H, Barrera JG, Wilson H, Opland DM, Faouzi MA, Gong Y, Jones JC, Rhodes CJ, Chua S Jr, Diano S, Horvath TL, Seeley RJ, Becker JB, Münzberg H, Myers MG Jr. Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding. Cell Metab 2009; 10(2): 89–98
doi: 10.1016/j.cmet.2009.06.011 pmid:19656487
83 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(2-3): 113–116
doi: 10.1016/0014-5793(96)00473-5 pmid:8674530
84 Ring LE, Zeltser LM. Disruption of hypothalamic leptin signaling in mice leads to early-onset obesity, but physiological adaptations in mature animals stabilize adiposity levels. J Clin Invest 2010; 120(8): 2931–2941
doi: 10.1172/JCI41985 pmid:20592471
85 Pan W, Hsuchou H, Jayaram B, Khan RS, Huang EY, Wu X, Chen C, Kastin AJ. Leptin action on nonneuronal cells in the CNS: potential clinical applications. Ann N Y Acad Sci 2012; 1264(1): 64–71
doi: 10.1111/j.1749-6632.2012.06472.x pmid:22530983
86 Morton GJ, Niswender KD, Rhodes CJ, Myers MG Jr, Blevins JE, Baskin DG, Schwartz MW. Arcuate nucleus-specific leptin receptor gene therapy attenuates the obesity phenotype of Koletsky (fa(k)/fa(k)) rats. Endocrinology 2003; 144(5): 2016–2024
doi: 10.1210/en.2002-0115 pmid:12697710
87 Myers MG Jr, Münzberg H, Leinninger GM, Leshan RL. The geometry of leptin action in the brain: more complicated than a simple ARC. Cell Metab 2009; 9(2): 117–123
doi: 10.1016/j.cmet.2008.12.001 pmid:19187770
88 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(6): 1375–1385
doi: 10.1016/S0896-6273(00)80656-X pmid:9883730
89 Balthasar N, Coppari R, McMinn J, Liu SM, Lee CE, Tang V, Kenny CD, McGovern RA, Chua SC Jr, Elmquist JK, Lowell BB. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 2004; 42(6): 983–991
doi: 10.1016/j.neuron.2004.06.004 pmid:15207242
90 Cowley MA, Smart JL, Rubinstein M, Cerdán 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(6836): 480–484
doi: 10.1038/35078085 pmid:11373681
91 Fan W, Boston BA, Kesterson RA, Hruby VJ, Cone RD. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 1997; 385(6612): 165–168
doi: 10.1038/385165a0 pmid:8990120
92 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(12): 2119–2123
doi: 10.2337/diabetes.46.12.2119 pmid:9392508
93 Hillebrand JJ, de Wied D, Adan RA. Neuropeptides, food intake and body weight regulation: a hypothalamic focus. Peptides 2002; 23(12): 2283–2306
doi: 10.1016/S0196-9781(02)00269-3 pmid:12535710
94 Butler AA, Cone RD. The melanocortin receptors: lessons from knockout models. Neuropeptides 2002; 36(2-3): 77–84
doi: 10.1054/npep.2002.0890 pmid:12359499
95 Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 1997; 88(1): 131–141
doi: 10.1016/S0092-8674(00)81865-6 pmid:9019399
96 Butler AA, Kesterson RA, Khong K, Cullen MJ, Pelleymounter MA, Dekoning J, Baetscher M, Cone RD. A unique metabolic syndrome causes obesity in the melanocortin-3 receptor-deficient mouse. Endocrinology 2000; 141(9): 3518–3521
doi: 10.1210/en.141.9.3518 pmid:10965927
97 Krude H, Biebermann H, Luck W, Horn R, Brabant G, Grüters A. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 1998; 19(2): 155–157
doi: 10.1038/509 pmid:9620771
98 Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, Lango Allen H, Lindgren CM, Luan J, M?gi R, Randall JC, Vedantam S, Winkler TW, Qi L, Workalemahu T, Heid IM, Steinthorsdottir V, Stringham HM, Weedon MN, Wheeler E, Wood AR, Ferreira T, Weyant RJ, Segrè AV, Estrada K, Liang L, Nemesh J, Park JH, Gustafsson S, Kilpel?inen TO, Yang J, Bouatia-Naji N, Esko T, Feitosa MF, Kutalik Z, Mangino M, Raychaudhuri S, Scherag A, Smith AV, Welch R, Zhao JH, Aben KK, Absher DM, Amin N, Dixon AL, Fisher E, Glazer NL, Goddard ME, Heard-Costa NL, Hoesel V, Hottenga JJ, Johansson A, Johnson T, Ketkar S, Lamina C, Li S, Moffatt MF, Myers RH, Narisu N, Perry JR, Peters MJ, Preuss M, Ripatti S, Rivadeneira F, Sandholt C, Scott LJ, Timpson NJ, Tyrer JP, van Wingerden S, Watanabe RM, White CC, Wiklund F, Barlassina C, Chasman DI, Cooper MN, Jansson JO, Lawrence RW, Pellikka N, Prokopenko I, Shi J, Thiering E, Alavere H, Alibrandi MT, Almgren P, Arnold AM, Aspelund T, Atwood LD, Balkau B, Balmforth AJ, Bennett AJ, Ben-Shlomo Y, Bergman RN, Bergmann S, Biebermann H, Blakemore AI, Boes T, Bonnycastle LL, Bornstein SR, Brown MJ, Buchanan TA, Busonero F, Campbell H, Cappuccio FP, Cavalcanti-Proen?a C, Chen YD, Chen CM, Chines PS, Clarke R, Coin L, Connell J, Day IN, den Heijer M, Duan J, Ebrahim S, Elliott P, Elosua R, Eiriksdottir G, Erdos MR, Eriksson JG, Facheris MF, Felix SB, Fischer-Posovszky P, Folsom AR, Friedrich N, Freimer NB, Fu M, Gaget S, Gejman PV, Geus EJ, Gieger C, Gjesing AP, Goel A, Goyette P, Grallert H, Gr?ssler J, Greenawalt DM, Groves CJ, Gudnason V, Guiducci C, Hartikainen AL, Hassanali N, Hall AS, Havulinna AS, Hayward C, Heath AC, Hengstenberg C, Hicks AA, Hinney A, Hofman A, Homuth G, Hui J, Igl W, Iribarren C, Isomaa B, Jacobs KB, Jarick I, Jewell E, John U, J?rgensen T, Jousilahti P, Jula A, Kaakinen M, Kajantie E, Kaplan LM, Kathiresan S, Kettunen J, Kinnunen L, Knowles JW, Kolcic I, K?nig IR, Koskinen S, Kovacs P, Kuusisto J, Kraft P, Kval?y K, Laitinen J, Lantieri O, Lanzani C, Launer LJ, Lecoeur C, Lehtim?ki T, Lettre G, Liu J, Lokki ML, Lorentzon M, Luben RN, Ludwig B, Manunta P, Marek D, Marre M, Martin NG, McArdle WL, McCarthy A, McKnight B, Meitinger T, Melander O, Meyre D, Midthjell K, Montgomery GW, Morken MA, Morris AP, Mulic R, Ngwa JS, Nelis M, Neville MJ, Nyholt DR, O’Donnell CJ, O’Rahilly S, Ong KK, Oostra B, Paré G, Parker AN, Perola M, Pichler I, Pietil?inen KH, Platou CG, Polasek O, Pouta A, Rafelt S, Raitakari O, Rayner NW, Ridderstr?le M, Rief W, Ruokonen A, Robertson NR, Rzehak P, Salomaa V, Sanders AR, Sandhu MS, Sanna S, Saramies J, Savolainen MJ, Scherag S, Schipf S, Schreiber S, Schunkert H, Silander K, Sinisalo J, Siscovick DS, Smit JH, Soranzo N, Sovio U, Stephens J, Surakka I, Swift AJ, Tammesoo ML, Tardif JC, Teder-Laving M, Teslovich TM, Thompson JR, Thomson B, T?njes A, Tuomi T, van Meurs JB, van Ommen GJ, Vatin V, Viikari J, Visvikis-Siest S, Vitart V, Vogel CI, Voight BF, Waite LL, Wallaschofski H, Walters GB, Widen E, Wiegand S, Wild SH, Willemsen G, Witte DR, Witteman JC, Xu J, Zhang Q, Zgaga L, Ziegler A, Zitting P, Beilby JP, Farooqi IS, Hebebrand J, Huikuri HV, James AL, K?h?nen M, Levinson DF, Macciardi F, Nieminen MS, Ohlsson C, Palmer LJ, Ridker PM, Stumvoll M, Beckmann JS, Boeing H, Boerwinkle E, Boomsma DI, Caulfield MJ, Chanock SJ, Collins FS, Cupples LA, Smith GD, Erdmann J, Froguel P, Gr?nberg H, Gyllensten U, Hall P, Hansen T, Harris TB, Hattersley AT, Hayes RB, Heinrich J, Hu FB, Hveem K, Illig T, Jarvelin MR, Kaprio J, Karpe F, Khaw KT, Kiemeney LA, Krude H, Laakso M, Lawlor DA, Metspalu A, Munroe PB, Ouwehand WH, Pedersen O, Penninx BW, Peters A, Pramstaller PP, Quertermous T, Reinehr T, Rissanen A, Rudan I, Samani NJ, Schwarz PE, Shuldiner AR, Spector TD, Tuomilehto J, Uda M, Uitterlinden A, Valle TT, Wabitsch M, Waeber G, Wareham NJ, Watkins H, Wilson JF, Wright AF, Zillikens MC, Chatterjee N, McCarroll SA, Purcell S, Schadt EE, Visscher PM, Assimes TL, Borecki IB, Deloukas P, Fox CS, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, Mohlke KL, O’Connell JR, Peltonen L, Schlessinger D, Strachan DP, van Duijn CM, Wichmann HE, Frayling TM, Thorsteinsdottir U, Abecasis GR, Barroso I, Boehnke M, Stefansson K, North KE, McCarthy MI, Hirschhorn JN, Ingelsson E, Loos RJ. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet 2010; 42(11): 937–948
doi: 10.1038/ng.686 pmid:20935630
99 Loos RJ, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP, Beckmann JS, Berndt SI, Jacobs KB, Chanock SJ, Hayes RB, Bergmann S, Bennett AJ, Bingham SA, Bochud M, Brown M, Cauchi S, Connell JM, Cooper C, Smith GD, Day I, Dina C, De S, Dermitzakis ET, Doney AS, Elliott KS, Elliott P, Evans DM, Sadaf Farooqi I, Froguel P, Ghori J, Groves CJ, Gwilliam R, Hadley D, Hall AS, Hattersley AT, Hebebrand J, Heid IM, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Herrera B, Hinney A, Hunt SE, Jarvelin MR, Johnson T, Jolley JD, Karpe F, Keniry A, Khaw KT, Luben RN, Mangino M, Marchini J, McArdle WL, McGinnis R, Meyre D, Munroe PB, Morris AD, Ness AR, Neville MJ, Nica AC, Ong KK, O’Rahilly S, Owen KR, Palmer CN, Papadakis K, Potter S, Pouta A, Qi L, Randall JC, Rayner NW, Ring SM, Sandhu MS, Scherag A, Sims MA, Song K, Soranzo N, Speliotes EK, Syddall HE, Teichmann SA, Timpson NJ, Tobias JH, Uda M, Vogel CI, Wallace C, Waterworth DM, Weedon MN, Willer CJ, Wraight, Yuan X, Zeggini E, Hirschhorn JN, Strachan DP, Ouwehand WH, Caulfield MJ, Samani NJ, Frayling TM, Vollenweider P, Waeber G, Mooser V, Deloukas P, McCarthy MI, Wareham NJ, Barroso I, Jacobs KB, Chanock SJ, Hayes RB, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Kraft P, Hankinson SE, Hunter DJ, Hu FB, Lyon HN, Voight BF, Ridderstrale M, Groop L, Scheet P, Sanna S, Abecasis GR, Albai G, Nagaraja R, Schlessinger D, Jackson AU, Tuomilehto J, Collins FS, Boehnke M, Mohlke KL. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet 2008; 40(6): 768–775
doi: 10.1038/ng.140 pmid:18454148
100 Hahn TM, Breininger JF, Baskin DG, Schwartz MW. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat Neurosci 1998; 1(4): 271–272
doi: 10.1038/1082 pmid:10195157
101 Luo N, Marcelin G, Liu SM, Schwartz G, Chua S Jr. Neuropeptide Y and agouti-related peptide mediate complementary functions of hyperphagia and reduced energy expenditure in leptin receptor deficiency. Endocrinology 2011; 152(3): 883–889
doi: 10.1210/en.2010-1135 pmid:21285324
102 Gropp E, Shanabrough M, Borok E, Xu AW, Janoschek R, Buch T, Plum L, Balthasar N, Hampel B, Waisman A, Barsh GS, Horvath TL, Brüning JC. Agouti-related peptide-expressing neurons are mandatory for feeding. Nat Neurosci 2005; 8(10): 1289–1291
doi: 10.1038/nn1548 pmid:16158063
103 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(5748): 683–685
doi: 10.1126/science.1115524 pmid:16254186
104 Bewick GA, Gardiner JV, Dhillo WS, Kent AS, White NE, Webster Z, Ghatei MA, Bloom SR. Post-embryonic ablation of AgRP neurons in mice leads to a lean, hypophagic phenotype. FASEB J 2005; 19(12): 1680–1682
pmid:16099943
105 Aponte Y, Atasoy D, Sternson SM. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat Neurosci 2011; 14(3): 351–355
doi: 10.1038/nn.2739 pmid:21209617
106 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(5335): 135–138
doi: 10.1126/science.278.5335.135 pmid:9311920
107 Qian S, Chen H, Weingarth D, Trumbauer ME, Novi DE, Guan X, Yu H, Shen Z, Feng Y, Frazier E, Chen A, Camacho RE, Shearman LP, Gopal-Truter S, MacNeil DJ, Van der Ploeg LH, Marsh DJ. Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Mol Cell Biol 2002; 22(14): 5027–5035
doi: 10.1128/MCB.22.14.5027-5035.2002 pmid:12077332
108 Tong Q, Ye CP, 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(9): 998–1000
doi: 10.1038/nn.2167 pmid:19160495
109 van de Wall E, Leshan R, Xu AW, Balthasar N, Coppari R, Liu SM, Jo YH, MacKenzie RG, Allison DB, Dun NJ, Elmquist J, Lowell BB, Barsh GS, de Luca C, Myers MG Jr, Schwartz GJ, Chua SC Jr. Collective and individual functions of leptin receptor modulated neurons controlling metabolism and ingestion. Endocrinology 2008; 149(4): 1773–1785
doi: 10.1210/en.2007-1132 pmid:18162515
110 Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, Tang V, Liu SM, Ludwig T, Chua SC Jr, Lowell BB, Elmquist JK. The hypothalamic arcuate nucleus: a key site for mediating leptin’s effects on glucose homeostasis and locomotor activity. Cell Metab 2005; 1(1): 63–72
doi: 10.1016/j.cmet.2004.12.004 pmid:16054045
111 Morton GJ, Gelling RW, Niswender KD, Morrison CD, Rhodes CJ, Schwartz MW. Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons. Cell Metab 2005; 2(6): 411–420
doi: 10.1016/j.cmet.2005.10.009 pmid:16330326
112 Vong L, Ye C, Yang Z, Choi B, Chua S Jr, Lowell BB. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 2011; 71(1): 142–154
doi: 10.1016/j.neuron.2011.05.028 pmid:21745644
113 Leshan RL, Greenwald-Yarnell M, Patterson CM, Gonzalez IE, Myers MG Jr. Leptin action through hypothalamic nitric oxide synthase-1-expressing neurons controls energy balance. Nat Med 2012; 18(5): 820–823
doi: 10.1038/nm.2724 pmid:22522563
114 Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bj?orbaek 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(1): 111–120
doi: 10.1172/JCI10741 pmid:11134186
115 Kim MS, Small CJ, Stanley SA, Morgan DG, Seal LJ, Kong WM, Edwards CM, Abusnana S, Sunter D, Ghatei MA, Bloom SR. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest 2000; 105(7): 1005–1011
doi: 10.1172/JCI8857 pmid:10749579
116 Balthasar N, Dalgaard LT, Lee CE, Yu J, Funahashi H, Williams T, Ferreira M, Tang V, McGovern RA, Kenny CD, Christiansen LM, Edelstein E, Choi B, Boss O, Aschkenasi C, Zhang CY, Mountjoy K, Kishi T, Elmquist JK, Lowell BB. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 2005; 123(3): 493–505
doi: 10.1016/j.cell.2005.08.035 pmid:16269339
117 Ghamari-Langroudi M, Srisai D, Cone RD. Multinodal regulation of the arcuate/paraventricular nucleus circuit by leptin. Proc Natl Acad Sci USA 2011; 108(1): 355–360
doi: 10.1073/pnas.1016785108 pmid:21169216
118 Xu B, Goulding EH, Zang K, Cepoi D, Cone RD, Jones KR, Tecott LH, Reichardt LF. Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nat Neurosci 2003; 6(7): 736–742
doi: 10.1038/nn1073 pmid:12796784
119 Sternson SM, Shepherd GM, Friedman JM. Topographic mapping of VMH → arcuate nucleus microcircuits and their reorganization by fasting. Nat Neurosci 2005; 8(10): 1356–1363
doi: 10.1038/nn1550 pmid:16172601
120 Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, Confavreux C, Klemenhagen KC, Tanaka KF, Gingrich JA, Guo XE, Tecott LH, Mann JJ, Hen R, Horvath TL, Karsenty G. A serotonin-dependent mechanism explains the leptin regulation of bone mass, appetite, and energy expenditure. Cell 2009; 138(5): 976–989
doi: 10.1016/j.cell.2009.06.051 pmid:19737523
121 Morris DL, Cho KW, Rui L. Critical role of the Src homology 2 (SH2) domain of neuronal SH2B1 in the regulation of body weight and glucose homeostasis in mice. Endocrinology 2010; 151(8): 3643–3651
doi: 10.1210/en.2010-0254 pmid:20484460
122 Coppari R, Bj?rb?k C. Leptin revisited: its mechanism of action and potential for treating diabetes. Nat Rev Drug Discov 2012; 11(9): 692–708
doi: 10.1038/nrd3757 pmid:22935803
123 Moraes-Vieira PM, Bassi EJ, Araujo RC, Camara NO. Leptin as a link between the immune system and kidney-related diseases: leading actor or just a coadjuvant? Obes Rev 2012; 13(8): 733–743
doi: 10.1111/j.1467-789X.2012.00997.x pmid:22498577
124 Ceccarini G, Flavell RR, Butelman ER, Synan M, Willnow TE, Bar-Dagan M, Goldsmith SJ, Kreek MJ, Kothari P, Vallabhajosula S, Muir TW, Friedman JM. PET imaging of leptin biodistribution and metabolism in rodents and primates. Cell Metab 2009; 10(2): 148–159
doi: 10.1016/j.cmet.2009.07.001 pmid:19656493
125 Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. Leptin enters the brain by a saturable system independent of insulin. Peptides 1996; 17(2): 305–311
doi: 10.1016/0196-9781(96)00025-3 pmid:8801538
126 Hileman SM, Pierroz DD, Masuzaki H, Bj?rbaek 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(3): 775–783
doi: 10.1210/en.143.3.775 pmid:11861497
127 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(12): 1449–1453
doi: 10.1016/S0196-9781(99)00156-4 pmid:10698121
128 Tu H, Kastin AJ, Hsuchou H, Pan W. Soluble receptor inhibits leptin transport. J Cell Physiol 2008; 214(2): 301–305
doi: 10.1002/jcp.21195 pmid:17620316
129 Dietrich MO, Spuch C, Antequera D, Rodal I, de Yébenes JG, Molina JA, Bermejo F, Carro E. Megalin mediates the transport of leptin across the blood-CSF barrier. Neurobiol Aging 2008; 29(6): 902–912
doi: 10.1016/j.neurobiolaging.2007.01.008 pmid:17324488
130 El-Haschimi K, Pierroz DD, Hileman SM, Bj?rbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest 2000; 105(12): 1827–1832
doi: 10.1172/JCI9842 pmid:10862798
131 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(9021): 159–161
doi: 10.1016/S0140-6736(96)03173-X pmid:8684156
132 Banks WA, DiPalma CR, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity. Peptides 1999; 20(11): 1341–1345
doi: 10.1016/S0196-9781(99)00139-4 pmid:10612449
133 Herde MK, Geist K, Campbell RE, Herbison AE. Gonadotropin-releasing hormone neurons extend complex highly branched dendritic trees outside the blood-brain barrier. Endocrinology 2011; 152(10): 3832–3841
doi: 10.1210/en.2011-1228 pmid:21791557
134 Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity is acquired and reversible. Am J Physiol Endocrinol Metab 2003; 285(1): E10–E15
pmid:12618361
135 Belouzard S, Delcroix D, Rouillé Y. Low levels of expression of leptin receptor at the cell surface result from constitutive endocytosis and intracellular retention in the biosynthetic pathway. J Biol Chem 2004; 279(27): 28499–28508
doi: 10.1074/jbc.M400508200 pmid:15123629
136 Diano S, Kalra SP, Horvath TL. Leptin receptor immunoreactivity is associated with the Golgi apparatus of hypothalamic neurons and glial cells. J Neuroendocrinol 1998; 10(9): 647–650
doi: 10.1046/j.1365-2826.1998.00261.x pmid:9744481
137 Seo S, Guo DF, Bugge K, Morgan DA, Rahmouni K, Sheffield VC. Requirement of Bardet-Biedl syndrome proteins for leptin receptor signaling. Hum Mol Genet 2009; 18(7): 1323–1331
doi: 10.1093/hmg/ddp031 pmid:19150989
138 Rahmouni K, Fath MA, Seo S, Thedens DR, Berry CJ, Weiss R, Nishimura DY, Sheffield VC. Leptin resistance contributes to obesity and hypertension in mouse models of Bardet-Biedl syndrome. J Clin Invest 2008; 118(4): 1458–1467
doi: 10.1172/JCI32357 pmid:18317593
139 Bj?rbaek 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(4): 619–625
doi: 10.1016/S1097-2765(00)80062-3 pmid:9660946
140 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(1-2): 109–118
doi: 10.1016/S0303-7207(02)00178-8 pmid:12354677
141 Loh K, Fukushima A, Zhang X, Galic S, Briggs D, Enriori PJ, Simonds S, Wiede F, Reichenbach A, Hauser C, Sims NA, Bence KK, Zhang S, Zhang ZY, Kahn BB, Neel BG, Andrews ZB, Cowley MA, Tiganis T. Elevated hypothalamic TCPTP in obesity contributes to cellular leptin resistance. Cell Metab 2011; 14(5): 684–699
doi: 10.1016/j.cmet.2011.09.011 pmid:22000926
142 Gamber KM, Huo L, Ha S, Hairston JE, Greeley S, Bjj?rb?k C. Over-expression of leptin receptors in hypothalamic POMC neurons increases susceptibility to diet-induced obesity. PLoS ONE 2012; 7(1): e30485
doi: 10.1371/journal.pone.0030485 pmid:22276206
143 Bj?rbaek C, El-Haschimi K, Frantz JD, Flier JS. The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 1999; 274(42): 30059–30065
doi: 10.1074/jbc.274.42.30059 pmid:10514492
144 Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bj?rbaek C, Flier JS. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med 2004; 10(7): 734–738
doi: 10.1038/nm1072 pmid:15220914
145 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(7): 739–743
doi: 10.1038/nm1071 pmid:15208705
146 Peralta S, Carrascosa JM, Gallardo N, Ros M, Arribas C. Ageing increases SOCS-3 expression in rat hypothalamus: effects of food restriction. Biochem Biophys Res Commun 2002; 296(2): 425–428
doi: 10.1016/S0006-291X(02)00906-3 pmid:12163036
147 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(4): 894–906
doi: 10.2337/db09-1024 pmid:20068134
148 Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, Haj F, Wang Y, Minokoshi Y, Kim YB, Elmquist JK, Tartaglia LA, Kahn BB, Neel BG. PTP1B regulates leptin signal transduction in vivo. Dev Cell 2002; 2(4): 489–495
doi: 10.1016/S1534-5807(02)00148-X pmid:11970898
149 Cheng A, Uetani N, Simoncic PD, Chaubey VP, Lee-Loy A, McGlade CJ, Kennedy BP, Tremblay ML. Attenuation of leptin action and regulation of obesity by protein tyrosine phosphatase 1B. Dev Cell 2002; 2(4): 497–503
doi: 10.1016/S1534-5807(02)00149-1 pmid:11970899
150 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(8): 917–924
doi: 10.1038/nm1435 pmid:16845389
151 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(9): 4227–4237
doi: 10.1210/en.2012-1548 pmid:22802463
152 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(3): 720–734
doi: 10.1172/JCI39620 pmid:20160350
153 Morrison CD, White CL, Wang Z, Lee SY, Lawrence DS, Cefalu WT, Zhang ZY, Gettys TW. Increased hypothalamic protein tyrosine phosphatase 1B contributes to leptin resistance with age. Endocrinology 2007; 148(1): 433–440
doi: 10.1210/en.2006-0672 pmid:17038557
154 White CL, Whittington A, Barnes MJ, Wang Z, Bray GA, Morrison CD. HF diets increase hypothalamic PTP1B and induce leptin resistance through both leptin-dependent and-independent mechanisms. Am J Physiol Endocrinol Metab 2009; 296(2): E291–E299
doi: 10.1152/ajpendo.90513.2008 pmid:19017730
155 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(5): 562–572
doi: 10.1016/j.cmet.2011.02.017 pmid:21531338
156 Plum L, Rother E, Münzberg H, Wunderlich FT, Morgan DA, Hampel B, Shanabrough M, Janoschek R, K?nner AC, Alber J, Suzuki A, Krone W, Horvath TL, Rahmouni K, Brüning JC. Enhanced leptin-stimulated PI3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab 2007; 6(6): 431–445
doi: 10.1016/j.cmet.2007.10.012 pmid:18054313
157 Plum L, Ma X, Hampel B, Balthasar N, Coppari R, Münzberg H, Shanabrough M, Burdakov D, Rother E, Janoschek R, Alber J, Belgardt BF, Koch L, Seibler J, Schwenk F, Fekete C, Suzuki A, Mak TW, Krone W, Horvath TL, Ashcroft FM, Brüning JC. Enhanced PIP3 signaling in POMC neurons causes KATP channel activation and leads to diet-sensitive obesity. J Clin Invest 2006; 116(7): 1886–1901
doi: 10.1172/JCI27123 pmid:16794735
158 Kl?ckener T, Hess S, Belgardt BF, Paeger L, Verhagen LA, Husch A, Sohn JW, Hampel B, Dhillon H, Zigman JM, Lowell BB, Williams KW, Elmquist JK, Horvath TL, Kloppenburg P, Brüning JC. High-fat feeding promotes obesity via insulin receptor/PI3K-dependent inhibition of SF-1 VMH neurons. Nat Neurosci 2011; 14(7): 911–918
doi: 10.1038/nn.2847 pmid:21642975
159 Li Z, Zhou Y, Carter-Su C, Myers MG Jr, Rui L. SH2B1 enhances leptin signaling by both Janus kinase 2 Tyr813 phosphorylation-dependent and-independent mechanisms. Mol Endocrinol 2007; 21(9): 2270–2281
doi: 10.1210/me.2007-0111 pmid:17565041
160 Rui L, Carter-Su C. Identification of SH2-bβ as a potent cytoplasmic activator of the tyrosine kinase Janus kinase 2. Proc Natl Acad Sci USA 1999; 96(13): 7172–7177
doi: 10.1073/pnas.96.13.7172 pmid:10377387
161 Morris DL, Cho KW, Zhou Y, Rui L. SH2B1 enhances insulin sensitivity by both stimulating the insulin receptor and inhibiting tyrosine dephosphorylation of insulin receptor substrate proteins. Diabetes 2009; 58(9): 2039–2047
doi: 10.2337/db08-1388 pmid:19542202
162 Song W, Ren D, Li W, Jiang L, Cho KW, Huang P, Fan C, Song Y, Liu Y, Rui L. SH2B regulation of growth, metabolism, and longevity in both insects and mammals. Cell Metab 2010; 11(5): 427–437
doi: 10.1016/j.cmet.2010.04.002 pmid:20417156
163 Jamshidi Y, Snieder H, Ge D, Spector TD, O’Dell SD. The SH2B gene is associated with serum leptin and body fat in normal female twins. Obesity (Silver Spring) 2007; 15(1): 5–9
doi: 10.1038/oby.2007.637 pmid:17228025
164 Renstr?m F, Payne F, Nordstr?m A, Brito EC, Rolandsson O, Hallmans G, Barroso I, Nordstr?m P, Franks PW. Replication and extension of genome-wide association study results for obesity in 4923 adults from northern Sweden. Hum Mol Genet 2009; 18(8): 1489–1496
doi: 10.1093/hmg/ddp041 pmid:19164386
165 Hester JM, Wing MR, Li J, Palmer ND, Xu J, Hicks PJ, Roh BH, Norris JM, Wagenknecht LE, Langefeld CD, Freedman BI, Bowden DW, Ng MC. Implication of European-derived adiposity loci in African Americans. Int J Obes (Lond) 2012; 36(3): 465–473
doi: 10.1038/ijo.2011.131 pmid:21750520
166 Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, Berndt SI, Elliott AL, Jackson AU, Lamina C, Lettre G, Lim N, Lyon HN, McCarroll SA, Papadakis K, Qi L, Randall JC, Roccasecca RM, Sanna S, Scheet P, Weedon MN, Wheeler E, Zhao JH, Jacobs LC, Prokopenko I, Soranzo N, Tanaka T, Timpson NJ, Almgren P, Bennett A, Bergman RN, Bingham SA, Bonnycastle LL, Brown M, Burtt NP, Chines P, Coin L, Collins FS, Connell JM, Cooper C, Smith GD, Dennison EM, Deodhar P, Elliott P, Erdos MR, Estrada K, Evans DM, Gianniny L, Gieger C, Gillson CJ, Guiducci C, Hackett R, Hadley D, Hall AS, Havulinna AS, Hebebrand J, Hofman A, Isomaa B, Jacobs KB, Johnson T, Jousilahti P, Jovanovic Z, Khaw KT, Kraft P, Kuokkanen M, Kuusisto J, Laitinen J, Lakatta EG, Luan J, Luben RN, Mangino M, McArdle WL, Meitinger T, Mulas A, Munroe PB, Narisu N, Ness AR, Northstone K, O’Rahilly S, Purmann C, Rees MG, Ridderstr?le M, Ring SM, Rivadeneira F, Ruokonen A, Sandhu MS, Saramies J, Scott LJ, Scuteri A, Silander K, Sims MA, Song K, Stephens J, Stevens S, Stringham HM, Tung YC, Valle TT, Van Duijn CM, Vimaleswaran KS, Vollenweider P, Waeber G, Wallace C, Watanabe RM, Waterworth DM, Watkins N, Witteman JC, Zeggini E, Zhai G, Zillikens MC, Altshuler D, Caulfield MJ, Chanock SJ, Farooqi IS, Ferrucci L, Guralnik JM, Hattersley AT, Hu FB, Jarvelin MR, Laakso M, Mooser V, Ong KK, Ouwehand WH, Salomaa V, Samani NJ, Spector TD, Tuomi T, Tuomilehto J, Uda M, Uitterlinden AG, Wareham NJ, Deloukas P, Frayling TM, Groop LC, Hayes RB, Hunter DJ, Mohlke KL, Peltonen L, Schlessinger D, Strachan DP, Wichmann HE, McCarthy MI, Boehnke M, Barroso I, Abecasis GR, Hirschhorn JN. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat Genet 2009; 41(1): 25–34
doi: 10.1038/ng.287 pmid:19079261
167 Thorleifsson G, Walters GB, Gudbjartsson DF, Steinthorsdottir V, Sulem P, Helgadottir A, Styrkarsdottir U, Gretarsdottir S, Thorlacius S, Jonsdottir I, Jonsdottir T, Olafsdottir EJ, Olafsdottir GH, Jonsson T, Jonsson F, Borch-Johnsen K, Hansen T, Andersen G, Jorgensen T, Lauritzen T, Aben KK, Verbeek AL, Roeleveld N, Kampman E, Yanek LR, Becker LC, Tryggvadottir L, Rafnar T, Becker DM, Gulcher J, Kiemeney LA, Pedersen O, Kong A, Thorsteinsdottir U, Stefansson K. Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat Genet 2009; 41(1): 18–24
doi: 10.1038/ng.274 pmid:19079260
168 Bochukova EG, Huang N, Keogh J, Henning E, Purmann C, Blaszczyk K, Saeed S, Hamilton-Shield J, Clayton-Smith J, O’Rahilly S, Hurles ME, Farooqi IS. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature 2010; 463(7281): 666–670
doi: 10.1038/nature08689 pmid:19966786
169 Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 2003; 348(12): 1085–1095
doi: 10.1056/NEJMoa022050 pmid:12646665
170 Lubrano-Berthelier C, Durand E, Dubern B, Shapiro A, Dazin P, Weill J, Ferron C, Froguel P, Vaisse C. Intracellular retention is a common characteristic of childhood obesity-associated MC4R mutations. Hum Mol Genet 2003; 12(2): 145–153
doi: 10.1093/hmg/ddg016 pmid:12499395
171 Marsh DJ, Hollopeter G, Huszar D, Laufer R, Yagaloff KA, Fisher SL, Burn P, Palmiter RD. Response of melanocortin-4 receptor-deficient mice to anorectic and orexigenic peptides. Nat Genet 1999; 21(1): 119–122
doi: 10.1038/5070 pmid:9916804
172 Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O’Rahilly S. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 1998; 20(2): 111–112
doi: 10.1038/2404 pmid:9771698
173 Pelleymounter MA, Cullen MJ, Wellman CL. Characteristics of BDNF-induced weight loss. Exp Neurol 1995; 131(2): 229–238
doi: 10.1016/0014-4886(95)90045-4 pmid:7534721
174 Yeo GS, Connie Hung CC, Rochford J, Keogh J, Gray J, Sivaramakrishnan S, O’Rahilly S, Farooqi IS. A de novo mutation affecting human TrkB associated with severe obesity and developmental delay. Nat Neurosci 2004; 7(11): 1187–1189
doi: 10.1038/nn1336 pmid:15494731
175 Liao GY, An JJ, Gharami K, Waterhouse EG, Vanevski F, Jones KR, Xu B. Dendritically targeted Bdnf mRNA is essential for energy balance and response to leptin. Nat Med 2012; 18(4): 564–571
doi: 10.1038/nm.2687 pmid:22426422
176 Knight ZA, Hannan KS, Greenberg ML, Friedman JM. Hyperleptinemia is required for the development of leptin resistance. PLoS ONE 2010; 5(6): e11376
doi: 10.1371/journal.pone.0011376 pmid:20613882
177 Ernst MB, Wunderlich CM, Hess S, Paehler M, Mesaros A, Koralov SB, Kleinridders A, Husch A, Münzberg H, Hampel B, Alber J, Kloppenburg P, Brüning JC, Wunderlich FT. Enhanced Stat3 activation in POMC neurons provokes negative feedback inhibition of leptin and insulin signaling in obesity. J Neurosci 2009; 29(37): 11582–11593
doi: 10.1523/JNEUROSCI.5712-08.2009 pmid:19759305
178 Tam J, Cinar R, Liu J, Godlewski G, Wesley D, Jourdan T, Szanda G, Mukhopadhyay B, Chedester L, Liow JS, Innis RB, Cheng K, Rice KC, Deschamps JR, Chorvat RJ, McElroy JF, Kunos G. Peripheral cannabinoid-1 receptor inverse agonism reduces obesity by reversing leptin resistance. Cell Metab 2012; 16(2): 167–179
doi: 10.1016/j.cmet.2012.07.002 pmid:22841573
179 Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 2011; 29(1): 415–445
doi: 10.1146/annurev-immunol-031210-101322 pmid:21219177
180 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(10): 4192–4199
doi: 10.1210/en.2004-1520 pmid:16002529
181 Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D. Hypothalamic IKKβ/NF-κB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008; 135(1): 61–73
doi: 10.1016/j.cell.2008.07.043 pmid:18854155
182 Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 2006; 116(11): 3015–3025
doi: 10.1172/JCI28898 pmid:17053832
183 Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, Tsukumo DM, Anhe G, Amaral ME, Takahashi HK, Curi R, Oliveira HC, Carvalheira JB, Bordin S, Saad MJ, Velloso LA. 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(2): 359–370
doi: 10.1523/JNEUROSCI.2760-08.2009 pmid:19144836
184 Kleinridders A, Schenten D, K?nner AC, Belgardt BF, Mauer J, Okamura T, Wunderlich FT, Medzhitov R, Brüning JC. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell Metab 2009; 10(4): 249–259
doi: 10.1016/j.cmet.2009.08.013 pmid:19808018
185 Hirosumi J, Tuncman G, Chang L, G?rgün CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS. A central role for JNK in obesity and insulin resistance. Nature 2002; 420(6913): 333–336
doi: 10.1038/nature01137 pmid:12447443
186 Sabio G, Cavanagh-Kyros J, Barrett T, Jung DY, Ko HJ, Ong H, Morel C, Mora A, Reilly J, Kim JK, Davis RJ. Role of the hypothalamic-pituitary-thyroid axis in metabolic regulation by JNK1. Genes Dev 2010; 24(3): 256–264
doi: 10.1101/gad.1878510 pmid:20080940
187 Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 2007; 8(7): 519–529
doi: 10.1038/nrm2199 pmid:17565364
188 Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 2010; 140(6): 900–917
doi: 10.1016/j.cell.2010.02.034 pmid:20303879
189 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(1): 35–51
doi: 10.1016/j.cmet.2008.12.004 pmid:19117545
190 Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii M, Ozawa K. Endoplasmic reticulum stress induces leptin resistance. Mol Pharmacol 2008; 74(6): 1610–1619
doi: 10.1124/mol.108.050070 pmid:18755873
191 Won JC, Jang PG, Namkoong C, Koh EH, Kim SK, Park JY, Lee KU, Kim MS. Central administration of an endoplasmic reticulum stress inducer inhibits the anorexigenic effects of leptin and insulin. Obesity (Silver Spring) 2009; 17(10): 1861–1865
doi: 10.1038/oby.2009.194 pmid:19543218
192 Ropelle ER, Flores MB, Cintra DE, Rocha GZ, Pauli JR, Morari J, de Souza CT, Moraes JC, Prada PO, Guadagnini D, Marin RM, Oliveira AG, Augusto TM, Carvalho HF, Velloso LA, Saad MJ, Carvalheira JB. IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKβ and ER stress inhibition. PLoS Biol 2010; 8(8): e1000465
doi: 10.1371/journal.pbio.1000465 pmid:20808781
193 Malik SA, Mari?o G, BenYounès A, Shen S, Harper F, Maiuri MC, Kroemer G. Neuroendocrine regulation of autophagy by leptin. Cell Cycle 2011; 10(17): 2917–2923
doi: 10.4161/cc.10.17.17067 pmid:21857156
194 Meng Q, Cai D. Defective hypothalamic autophagy directs the central pathogenesis of obesity via the IκB kinase beta (IKKβ)/NF-κB pathway. J Biol Chem 2011; 286(37): 32324–32332
doi: 10.1074/jbc.M111.254417 pmid:21784844
195 Kaushik S, Rodriguez-Navarro JA, Arias E, Kiffin R, Sahu S, Schwartz GJ, Cuervo AM, Singh R. Autophagy in hypothalamic AgRP neurons regulates food intake and energy balance. Cell Metab 2011; 14(2): 173–183
doi: 10.1016/j.cmet.2011.06.008 pmid:21803288
196 Quan W, Kim HK, Moon EY, Kim SS, Choi CS, Komatsu M, Jeong YT, Lee MK, Kim KW, Kim MS, Lee MS. Role of hypothalamic proopiomelanocortin neuron autophagy in the control of appetite and leptin response. Endocrinology 2012; 153(4): 1817–1826
doi: 10.1210/en.2011-1882 pmid:22334718
[1] So Jung Yang, Hun-Sung Kim, Kun-Ho Yoon. Analyzing the distinguishing factors that affect childhood obesity in South Korea[J]. Front. Med., 2018, 12(6): 707-716.
[2] Eun Young Lee, Kun-Ho Yoon. Epidemic obesity in children and adolescents: risk factors and prevention[J]. Front. Med., 2018, 12(6): 658-666.
[3] Ruiting Han, Junli Ma, Houkai Li. Mechanistic and therapeutic advances in non-alcoholic fatty liver disease by targeting the gut microbiota[J]. Front. Med., 2018, 12(6): 645-657.
[4] Tiange Wang, Min Xu, Yufang Bi, Guang Ning. Interplay between diet and genetic susceptibility in obesity and related traits[J]. Front. Med., 2018, 12(6): 601-607.
[5] Meng Dong, Jun Lin, Wonchung Lim, Wanzhu Jin, Hyuek Jong Lee. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia[J]. Front. Med., 2018, 12(2): 130-138.
[6] Tianhua Xu, Zitong Sheng, Li Yao. Obesity-related glomerulopathy: pathogenesis, pathologic, clinical characteristics and treatment[J]. Front. Med., 2017, 11(3): 340-348.
[7] Rahim Ullah, Yan Su, Yi Shen, Chunlu Li, Xiaoqin Xu, Jianwei Zhang, Ke Huang, Naveed Rauf, Yang He, Jingjing Cheng, Huaping Qin, Yu-Dong Zhou, Junfen Fu. Postnatal feeding with high-fat diet induces obesity and precocious puberty in C57BL/6J mouse pups: a novel model of obesity and puberty[J]. Front. Med., 2017, 11(2): 266-276.
[8] Lixia Gan,Wei Xiang,Bin Xie,Liqing Yu. Molecular mechanisms of fatty liver in obesity[J]. Front. Med., 2015, 9(3): 275-287.
[9] Shuwen Qian,Haiyan Huang,Qiqun Tang. Brown and beige fat: the metabolic function, induction, and therapeutic potential[J]. Front. Med., 2015, 9(2): 162-172.
[10] Jianping Ye. Beneficial metabolic activities of inflammatory cytokine interleukin 15 in obesity and type 2 diabetes[J]. Front. Med., 2015, 9(2): 139-145.
[11] Tao Wang,Weiping Jia,Cheng Hu. Advancement in genetic variants conferring obesity susceptibility from genome-wide association studies[J]. Front. Med., 2015, 9(2): 146-161.
[12] Jichun Yang, Jihong Kang, Youfei Guan. The mechanisms linking adiposopathy to type 2 diabetes[J]. Front Med, 2013, 7(4): 433-444.
[13] Xinjian Li, Jiying Xu, Haihong Yao, Yanfei Guo, Minna Chen, Wei Lu. Obesity and overweight prevalence and its association with undiagnosed hypertension in Shanghai population, China: a cross-sectional population-based survey[J]. Front Med, 2012, 6(3): 322-328.
[14] Ling-Yan XU PhD, Xin-Ran MA PhD, Xiao-Ying LI PhD, MD, Shu WANG PhD, Guang NING PhD, MD, Jie-Li LI PhD, Jian-Ming XU PhD, . Ablation of steroid receptor coactivator-3 in mice impairs adipogenesis and enhances energy expenditure[J]. Front. Med., 2010, 4(2): 229-234.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed