메뉴 건너뛰기




Volumn 34, Issue 3, 2013, Pages 309-338

The role of estrogens in control of energy balance and glucose homeostasis

Author keywords

[No Author keywords available]

Indexed keywords

ANASTROZOLE; CONJUGATED ESTROGEN; ESTRADIOL; ESTROGEN; ESTROGEN RECEPTOR ALPHA; ESTROGEN RECEPTOR BETA; ESTROGEN SULFOTRANSFERASE; EXEMESTANE; GESTAGEN; GLUCOSE TRANSPORTER 4; GSK 232802A; INSULIN; LEPTIN; LETROZOLE; NEUROPEPTIDE Y; PLACEBO; PROGESTERONE; RALOXIFENE; SELECTIVE ESTROGEN RECEPTOR MODULATOR; TAMOXIFEN; UNCLASSIFIED DRUG;

EID: 84878632677     PISSN: 0163769X     EISSN: None     Source Type: Journal    
DOI: 10.1210/er.2012-1055     Document Type: Review
Times cited : (921)

References (357)
  • 2
    • 0016826567 scopus 로고
    • Increased risk of endometrial carcinoma among users of conjugated estrogens
    • Ziel HK, Finkle WD. Increased risk of endometrial carcinoma among users of conjugated estrogens.NEngl J Med. 1975;293:1167-1170.
    • (1975) NEngl J Med. , vol.293 , pp. 1167-1170
    • Ziel, H.K.1    Finkle, W.D.2
  • 4
    • 0037125379 scopus 로고    scopus 로고
    • Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial
    • Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA. 2002;288: 321-333.
    • (2002) JAMA. , vol.288 , pp. 321-333
    • Rossouw, J.E.1    Anderson, G.L.2    Prentice, R.L.3
  • 5
    • 77955868505 scopus 로고    scopus 로고
    • Postmenopausal hormone therapy: an Endocrine Society scientific statement
    • Santen RJ, Allred DC, Ardoin SP, et al. Postmenopausal hormone therapy: an Endocrine Society scientific statement. J Clin Endocrinol Metab. 2010;95(7 suppl 1): s1-s66.
    • (2010) J Clin Endocrinol Metab. , vol.95 , Issue.7 SUPPL. 1
    • Santen, R.J.1    Allred, D.C.2    Ardoin, S.P.3
  • 6
    • 0038645816 scopus 로고    scopus 로고
    • The emergence of the metabolic syndrome with menopause
    • Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab. 2003;88:2404-2411.
    • (2003) J Clin Endocrinol Metab. , vol.88 , pp. 2404-2411
    • Carr, M.C.1
  • 7
    • 78650521925 scopus 로고    scopus 로고
    • Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity
    • Mauvais-Jarvis F. Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity. Trends Endocrinol Metab. 2011;22:24-33.
    • (2011) Trends Endocrinol Metab. , vol.22 , pp. 24-33
    • Mauvais-Jarvis, F.1
  • 8
    • 18844461988 scopus 로고    scopus 로고
    • Estrogen-the good, the bad, and the unexpected
    • Simpson ER, Misso M, Hewitt KN, et al. Estrogen-the good, the bad, and the unexpected. Endocr Rev. 2005;26: 322-330.
    • (2005) Endocr Rev. , vol.26 , pp. 322-330
    • Simpson, E.R.1    Misso, M.2    Hewitt, K.N.3
  • 9
    • 84861919844 scopus 로고    scopus 로고
    • Estrogen sulfotransferase: intracrinology meets metabolic diseases
    • Mauvais-Jarvis F. Estrogen sulfotransferase: intracrinology meets metabolic diseases. Diabetes. 2012;61:1353-1354.
    • (2012) Diabetes. , vol.61 , pp. 1353-1354
    • Mauvais-Jarvis, F.1
  • 10
    • 0030446327 scopus 로고    scopus 로고
    • Steroid sulfotransferases
    • Strott CA. Steroid sulfotransferases. Endocr Rev. 1996; 17:670-697.
    • (1996) Endocr Rev. , vol.17 , pp. 670-697
    • Strott, C.A.1
  • 11
    • 0015230827 scopus 로고
    • Mechanisms of action of steroid hormones
    • O'Malley BW. Mechanisms of action of steroid hormones. N Engl J Med. 1971;284:370-377.
    • (1971) N Engl J Med. , vol.284 , pp. 370-377
    • O'Malley, B.W.1
  • 12
  • 13
    • 57749178899 scopus 로고    scopus 로고
    • Non-classical genomic estrogen receptor (ER)/specificity protein and ER/activating protein-1 signaling pathways
    • Safe S, Kim K. Non-classical genomic estrogen receptor (ER)/specificity protein and ER/activating protein-1 signaling pathways. J Mol Endocrinol. 2008;41:263-275.
    • (2008) J Mol Endocrinol. , vol.41 , pp. 263-275
    • Safe, S.1    Kim, K.2
  • 14
    • 0030801841 scopus 로고    scopus 로고
    • Differential ligand activation of estrogen receptors ERα and ERβ at AP1 sites
    • Paech K, Webb P, Kuiper GG, et al. Differential ligand activation of estrogen receptors ERα and ERβ at AP1 sites. Science. 1997;277:1508-1510.
    • (1997) Science. , vol.277 , pp. 1508-1510
    • Paech, K.1    Webb, P.2    Kuiper, G.G.3
  • 15
    • 57749105457 scopus 로고    scopus 로고
    • Intermolecular interactions identify ligand-selective activity of estrogen receptor α/β dimers
    • Powell E, Xu W. Intermolecular interactions identify ligand-selective activity of estrogen receptor α/β dimers. Proc Natl Acad Sci USA. 2008;105:19012-19017.
    • (2008) Proc Natl Acad Sci USA. , vol.105 , pp. 19012-19017
    • Powell, E.1    Xu, W.2
  • 16
    • 73549104043 scopus 로고    scopus 로고
    • Genome-wide dynamics of chromatin binding of estrogen receptors α and β: mutual restriction and competitive site selection
    • Charn TH, Liu ET, Chang EC, Lee YK, Katzenellenbogen JA, Katzenellenbogen BS. Genome-wide dynamics of chromatin binding of estrogen receptors α and β: mutual restriction and competitive site selection. Mol Endocrinol. 2010;24:47-59.
    • (2010) Mol Endocrinol. , vol.24 , pp. 47-59
    • Charn, T.H.1    Liu, E.T.2    Chang, E.C.3    Lee, Y.K.4    Katzenellenbogen, J.A.5    Katzenellenbogen, B.S.6
  • 17
    • 0033637703 scopus 로고    scopus 로고
    • Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription
    • Shang Y,HuX, DiRenzo J, LazarMA,Brown M. Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell. 2000;103:843-852.
    • (2000) Cell. , vol.103 , pp. 843-852
    • Shang, Y.1    Hu, X.2    DiRenzo, J.3    Lazar, M.A.4    Brown, M.5
  • 18
    • 77149165088 scopus 로고    scopus 로고
    • Minireview: estrogenic protection of β-cell failure in metabolic diseases
    • Liu S, Mauvais-Jarvis F. Minireview: estrogenic protection of β-cell failure in metabolic diseases. Endocrinology. 2010;151:859-864.
    • (2010) Endocrinology. , vol.151 , pp. 859-864
    • Liu, S.1    Mauvais-Jarvis, F.2
  • 19
    • 37349081370 scopus 로고    scopus 로고
    • Extranuclear steroid receptors: nature and actions
    • Hammes SR, Levin ER. Extranuclear steroid receptors: nature and actions. Endocr Rev. 2007;28:726-741.
    • (2007) Endocr Rev. , vol.28 , pp. 726-741
    • Hammes, S.R.1    Levin, E.R.2
  • 20
    • 84861335162 scopus 로고    scopus 로고
    • Importance of oestrogen receptors to preserve functional β-cell mass in diabetes
    • Tiano JP, Mauvais-Jarvis F. Importance of oestrogen receptors to preserve functional β-cell mass in diabetes. Nat Rev Endocrinol. 2012;8:342-351.
    • (2012) Nat Rev Endocrinol. , vol.8 , pp. 342-351
    • Tiano, J.P.1    Mauvais-Jarvis, F.2
  • 21
    • 78650979007 scopus 로고    scopus 로고
    • The impact of an aromatase inhibitor on body composition and gonadal hormone levels in women with breast cancer
    • van Londen GJ, Perera S, Vujevich K, et al. The impact of an aromatase inhibitor on body composition and gonadal hormone levels in women with breast cancer. Breast Cancer Res Treat. 2011;125:441-446.
    • (2011) Breast Cancer Res Treat. , vol.125 , pp. 441-446
    • van Londen, G.J.1    Perera, S.2    Vujevich, K.3
  • 22
    • 78751683777 scopus 로고    scopus 로고
    • Genomic collaboration of estrogen receptor α and extracellular signal-regulated kinase 2 in regulating gene and proliferation programs
    • Madak-Erdogan Z, Lupien M, Stossi F, Brown M, Katzenellenbogen BS. Genomic collaboration of estrogen receptor α and extracellular signal-regulated kinase 2 in regulating gene and proliferation programs. Mol Cell Biol. 2011;31:226-236.
    • (2011) Mol Cell Biol. , vol.31 , pp. 226-236
    • Madak-Erdogan, Z.1    Lupien, M.2    Stossi, F.3    Brown, M.4    Katzenellenbogen, B.S.5
  • 23
    • 77950407928 scopus 로고    scopus 로고
    • Involvement of estrogen receptor variant ER-α36, not GPR30, in nongenomic estrogen signaling
    • Kang L, Zhang X, Xie Y, et al. Involvement of estrogen receptor variant ER-α36, not GPR30, in nongenomic estrogen signaling. Mol Endocrinol. 2010;24:709-721.
    • (2010) Mol Endocrinol. , vol.24 , pp. 709-721
    • Kang, L.1    Zhang, X.2    Xie, Y.3
  • 24
    • 0031451570 scopus 로고    scopus 로고
    • Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor
    • Laudet V. Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor. J Mol Endocrinol. 1997;19:207-226.
    • (1997) J Mol Endocrinol. , vol.19 , pp. 207-226
    • Laudet, V.1
  • 25
    • 0141431993 scopus 로고    scopus 로고
    • Resurrecting the ancestral steroid receptor: ancient origin of estrogen signaling
    • Thornton JW, Need E, Crews D. Resurrecting the ancestral steroid receptor: ancient origin of estrogen signaling. Science. 2003;301:1714-1717.
    • (2003) Science. , vol.301 , pp. 1714-1717
    • Thornton, J.W.1    Need, E.2    Crews, D.3
  • 26
    • 79951672289 scopus 로고    scopus 로고
    • Evolution of steroid receptors from an estrogen-sensitive ancestral receptor
    • Eick GN, Thornton JW. Evolution of steroid receptors from an estrogen-sensitive ancestral receptor. Mol Cell Endocrinol. 2011;334:31-38.
    • (2011) Mol Cell Endocrinol. , vol.334 , pp. 31-38
    • Eick, G.N.1    Thornton, J.W.2
  • 27
    • 33645691679 scopus 로고    scopus 로고
    • Evolution of hormone-receptor complexity by molecular exploitation
    • Bridgham JT, Carroll SM, Thornton JW. Evolution of hormone-receptor complexity by molecular exploitation. Science. 2006;312:97-101.
    • (2006) Science. , vol.312 , pp. 97-101
    • Bridgham, J.T.1    Carroll, S.M.2    Thornton, J.W.3
  • 28
    • 56449119335 scopus 로고    scopus 로고
    • Evolution of hormone signaling in elasmobranchs by exploitation of promiscuous receptors
    • Carroll SM, Bridgham JT, Thornton JW. Evolution of hormone signaling in elasmobranchs by exploitation of promiscuous receptors. Mol Biol Evol. 2008;25:2643-2652.
    • (2008) Mol Biol Evol. , vol.25 , pp. 2643-2652
    • Carroll, S.M.1    Bridgham, J.T.2    Thornton, J.W.3
  • 30
    • 0017189871 scopus 로고
    • Ovarian influences on the meal patterns of female rats
    • Blaustein JD, Wade GN. Ovarian influences on the meal patterns of female rats. Physiol Behav. 1976;17:201-208. 31. Drewett RF. Sexual behaviour and sexual motivation in the female rat. Nature. 1973;242:476-477.
    • (1976) Physiol Behav. , vol.17 , pp. 201-208
    • Blaustein, J.D.1    Wade, G.N.2
  • 31
    • 0015821522 scopus 로고
    • Sexual behaviour and sexual motivation in the female rat
    • Drewett RF. Sexual behaviour and sexual motivation in the female rat. Nature. 1973;242:476-477.
    • (1973) Nature. , vol.242 , pp. 476-477
    • Drewett, R.F.1
  • 32
    • 0034843320 scopus 로고    scopus 로고
    • Sex hormones and the selective estrogen receptor modulator tamoxifen modulate weekly body weights and food intakes in adolescent and adult rats
    • Wallen WJ, Belanger MP, Wittnich C. Sex hormones and the selective estrogen receptor modulator tamoxifen modulate weekly body weights and food intakes in adolescent and adult rats. J Nutr. 2001;131:2351-2357.
    • (2001) J Nutr. , vol.131 , pp. 2351-2357
    • Wallen, W.J.1    Belanger, M.P.2    Wittnich, C.3
  • 33
    • 0034751475 scopus 로고    scopus 로고
    • Def icits in E2-dependent control of feeding, weight gain, and cholecystokinin satiation in ER-α null mice
    • Geary N, Asarian L, Korach KS, Pfaff DW, Ogawa S. Def icits in E2-dependent control of feeding, weight gain, and cholecystokinin satiation in ER-α null mice. Endocrinology. 2001;142:4751-4757.
    • (2001) Endocrinology. , vol.142 , pp. 4751-4757
    • Geary, N.1    Asarian, L.2    Korach, K.S.3    Pfaff, D.W.4    Ogawa, S.5
  • 34
    • 33846046623 scopus 로고    scopus 로고
    • Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals
    • Gao Q, Mezei G, Nie Y, et al. Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nat Med. 2007; 13:89-94.
    • (2007) Nat Med. , vol.13 , pp. 89-94
    • Gao, Q.1    Mezei, G.2    Nie, Y.3
  • 35
    • 70349235444 scopus 로고    scopus 로고
    • Sex hormone-binding globulin and risk of type 2 diabetes in women and men
    • Ding EL, Song Y, Manson JE, et al. Sex hormone-binding globulin and risk of type 2 diabetes in women and men. N Engl J Med. 2009;361:1152-1163.
    • (2009) N Engl J Med. , vol.361 , pp. 1152-1163
    • Ding, E.L.1    Song, Y.2    Manson, J.E.3
  • 36
    • 12944276957 scopus 로고    scopus 로고
    • Aromatasedeficient (ArKO) mice have a phenotype of increased adiposity
    • Jones ME, Thorburn AW, Britt KL, et al. Aromatasedeficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci USA. 2000;97:12735-12740.
    • (2000) Proc Natl Acad Sci USA. , vol.97 , pp. 12735-12740
    • Jones, M.E.1    Thorburn, A.W.2    Britt, K.L.3
  • 38
    • 74949093473 scopus 로고    scopus 로고
    • Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERα deficient mice
    • Ribas V, Nguyen MT, Henstridge DC, et al. Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERα deficient mice. Am J Physiol Endocrinol Metab. 2010;298:E304-E319.
    • (2010) Am J Physiol Endocrinol Metab. , vol.298
    • Ribas, V.1    Nguyen, M.T.2    Henstridge, D.C.3
  • 39
    • 66449086930 scopus 로고    scopus 로고
    • Reduced energy expenditure and increased inflammation are early events in the development of ovariectomy-induced obesity
    • Rogers NH, Perfield JW II, Strissel KJ, Obin MS, Greenberg AS. Reduced energy expenditure and increased inflammation are early events in the development of ovariectomy-induced obesity. Endocrinology. 2009;150: 2161-2168.
    • (2009) Endocrinology. , vol.150 , pp. 2161-2168
    • Rogers, N.H.1    Perfield, J.W.2    Strissel, K.J.3    Obin, M.S.4    Greenberg, A.S.5
  • 40
    • 0033782039 scopus 로고    scopus 로고
    • The controls of eating: a shift from nutritional homeostasis to behavioral neuroscience
    • Smith GP. The controls of eating: a shift from nutritional homeostasis to behavioral neuroscience. Nutrition. 2000; 16:814-820.
    • (2000) Nutrition. , vol.16 , pp. 814-820
    • Smith, G.P.1
  • 41
    • 0019142594 scopus 로고
    • Neuroendocrinology of hyperphagias and obesities
    • Louis-Sylvestre J. Neuroendocrinology of hyperphagias and obesities. Reprod Nutr Dev. 1980;20:1545-1562.
    • (1980) Reprod Nutr Dev. , vol.20 , pp. 1545-1562
    • Louis-Sylvestre, J.1
  • 42
    • 0018865746 scopus 로고
    • Cortical activity and sleep in the rat lateral hypothalamic syndrome
    • Danguir J, Nicolaidis S. Cortical activity and sleep in the rat lateral hypothalamic syndrome. Brain Res. 1980;185: 305-321.
    • (1980) Brain Res. , vol.185 , pp. 305-321
    • Danguir, J.1    Nicolaidis, S.2
  • 43
    • 0019065207 scopus 로고
    • Effect of lateral hypothalamic lesions on regulation of body weight and adiposity in rats
    • Milam KM, Stern JS, Storlien LH, Keesey RE. Effect of lateral hypothalamic lesions on regulation of body weight and adiposity in rats. Am J Physiol. 1980;239:R337-R343.
    • (1980) Am J Physiol. , vol.239
    • Milam, K.M.1    Stern, J.S.2    Storlien, L.H.3    Keesey, R.E.4
  • 44
    • 0025232316 scopus 로고
    • Distribution of androgen and estrogen receptor mRNAcontaining cells in the rat brain: an in situ hybridization study
    • Simerly RB, Chang C, Muramatsu M, Swanson LW. Distribution of androgen and estrogen receptor mRNAcontaining cells in the rat brain: an in situ hybridization study. J Comp Neurol. 1990;294:76-95.
    • (1990) J Comp Neurol. , vol.294 , pp. 76-95
    • Simerly, R.B.1    Chang, C.2    Muramatsu, M.3    Swanson, L.W.4
  • 45
    • 0030669205 scopus 로고    scopus 로고
    • Differential expression of estrogen receptorα andβ immunoreactivity by oxytocin neurons of rat paraventricular nucleus
    • Simonian SX, Herbison AE. Differential expression of estrogen receptorα andβ immunoreactivity by oxytocin neurons of rat paraventricular nucleus. J Neuroendocrinol. 1997;9:803-806.
    • (1997) J Neuroendocrinol. , vol.9 , pp. 803-806
    • Simonian, S.X.1    Herbison, A.E.2
  • 46
    • 0030976010 scopus 로고    scopus 로고
    • Identification of estrogen receptor-containing neurons projecting to the rat supraoptic nucleus
    • Voisin DL, Simonian SX, Herbison AE. Identification of estrogen receptor-containing neurons projecting to the rat supraoptic nucleus. Neuroscience. 1997;78:215-228.
    • (1997) Neuroscience. , vol.78 , pp. 215-228
    • Voisin, D.L.1    Simonian, S.X.2    Herbison, A.E.3
  • 47
    • 0031910162 scopus 로고    scopus 로고
    • Differential distribution and regulation of estrogen receptor-α and-β mRNA within the female rat brain
    • Osterlund M, Kuiper GG, Gustafsson JA, Hurd YL. Differential distribution and regulation of estrogen receptor-α and-β mRNA within the female rat brain. Brain Res Mol Brain Res. 1998;54:175-180.
    • (1998) Brain Res Mol Brain Res. , vol.54 , pp. 175-180
    • Osterlund, M.1    Kuiper, G.G.2    Gustafsson, J.A.3    Hurd, Y.L.4
  • 48
    • 0036217363 scopus 로고    scopus 로고
    • Identification and characterization of a functionally distinct form of human estrogen receptor β
    • Wilkinson HA, Dahllund J, Liu H, et al. Identification and characterization of a functionally distinct form of human estrogen receptor β. Endocrinology. 2002;143:1558-1561.
    • (2002) Endocrinology. , vol.143 , pp. 1558-1561
    • Wilkinson, H.A.1    Dahllund, J.2    Liu, H.3
  • 49
    • 0041915552 scopus 로고    scopus 로고
    • Distribution of estrogen receptor β mRNA-containing cells in ovariectomized and estrogen-treated female rat brain
    • Shima N, Yamaguchi Y, Yuri K. Distribution of estrogen receptor β mRNA-containing cells in ovariectomized and estrogen-treated female rat brain. Anat Sci Int. 2003;78: 85-97.
    • (2003) Anat Sci Int. , vol.78 , pp. 85-97
    • Shima, N.1    Yamaguchi, Y.2    Yuri, K.3
  • 50
    • 2342584831 scopus 로고    scopus 로고
    • Distribution of estrogen receptor α and β in the mouse central nervous system: in vivo autoradiographic and immunocytochemical analyses
    • Merchenthaler I, Lane MV, Numan S, Dellovade TL. Distribution of estrogen receptor α and β in the mouse central nervous system: in vivo autoradiographic and immunocytochemical analyses. J Comp Neurol. 2004; 473:270-291.
    • (2004) J Comp Neurol. , vol.473 , pp. 270-291
    • Merchenthaler, I.1    Lane, M.V.2    Numan, S.3    Dellovade, T.L.4
  • 51
    • 0034735999 scopus 로고    scopus 로고
    • Obesity and disturbed lipoprotein profile in estrogen receptor-α-deficient male mice
    • Ohlsson C, Hellberg N, Parini P, et al. Obesity and disturbed lipoprotein profile in estrogen receptor-α-deficient male mice. Biochem Biophys Res Commun. 2000;278: 640-645.
    • (2000) Biochem Biophys Res Commun. , vol.278 , pp. 640-645
    • Ohlsson, C.1    Hellberg, N.2    Parini, P.3
  • 52
    • 29144482697 scopus 로고    scopus 로고
    • Effects of selective estrogen receptor agonists on food intake and body weight gain in rats
    • Roesch DM. Effects of selective estrogen receptor agonists on food intake and body weight gain in rats. Physiol Behav. 2006;87:39-44.
    • (2006) Physiol Behav. , vol.87 , pp. 39-44
    • Roesch, D.M.1
  • 53
    • 36849022711 scopus 로고    scopus 로고
    • Acute activation of ER α decreases food intake, meal size, and body weight in ovariectomized rats
    • Santollo J, Wiley MD, Eckel LA. Acute activation of ER α decreases food intake, meal size, and body weight in ovariectomized rats. Am J Physiol Regul Integr Comp Physiol. 2007;293:R2194-R2201.
    • (2007) Am J Physiol Regul Integr Comp Physiol. , vol.293
    • Santollo, J.1    Wiley, M.D.2    Eckel, L.A.3
  • 54
    • 0036346098 scopus 로고    scopus 로고
    • Estrogen receptor β is involved in the anorectic action of estrogen
    • Liang YQ, Akishita M, Kim S, et al. Estrogen receptor β is involved in the anorectic action of estrogen. Int J Obes Relat Metab Disord. 2002;26:1103-1109.
    • (2002) Int J Obes Relat Metab Disord. , vol.26 , pp. 1103-1109
    • Liang, Y.Q.1    Akishita, M.2    Kim, S.3
  • 55
    • 77957768699 scopus 로고    scopus 로고
    • Estrogen receptor-β selective ligands alleviate high-fat diet-and ovariectomy-induced obesity in mice
    • Yepuru M, Eswaraka J, Kearbey JD, et al. Estrogen receptor-β selective ligands alleviate high-fat diet-and ovariectomy-induced obesity in mice. J Biol Chem. 2010;285: 31292-31303.
    • (2010) J Biol Chem. , vol.285 , pp. 31292-31303
    • Yepuru, M.1    Eswaraka, J.2    Kearbey, J.D.3
  • 56
    • 77954244922 scopus 로고    scopus 로고
    • A critical review of fundamental controversies in the field of GPR30 research
    • Langer G, Bader B, Meoli L, et al. A critical review of fundamental controversies in the field of GPR30 research. Steroids. 2010;75:603-610.
    • (2010) Steroids. , vol.75 , pp. 603-610
    • Langer, G.1    Bader, B.2    Meoli, L.3
  • 57
    • 79951671672 scopus 로고    scopus 로고
    • Estrogen inhibits NPY secretion through membrane-associated estrogen receptor (ER)-α in clonal, immortalized hypothalamic neurons
    • Dhillon SS, Belsham DD. Estrogen inhibits NPY secretion through membrane-associated estrogen receptor (ER)-α in clonal, immortalized hypothalamic neurons. Int J Obes (Lond). 2011;35:198-207.
    • (2011) Int J Obes (Lond). , vol.35 , pp. 198-207
    • Dhillon, S.S.1    Belsham, D.D.2
  • 58
    • 33744970064 scopus 로고    scopus 로고
    • A G-protein-coupled estrogen receptor is involved in hypothalamic control of energy homeostasis
    • Qiu J, Bosch MA, Tobias SC, et al. A G-protein-coupled estrogen receptor is involved in hypothalamic control of energy homeostasis. J Neurosci. 2006;26:5649-5655.
    • (2006) J Neurosci. , vol.26 , pp. 5649-5655
    • Qiu, J.1    Bosch, M.A.2    Tobias, S.C.3
  • 59
    • 77957260252 scopus 로고    scopus 로고
    • Contribution of a membrane estrogen receptor to the estrogenic regulation of body temperature and energy homeostasis
    • Roepke TA, Bosch MA, Rick EA, et al. Contribution of a membrane estrogen receptor to the estrogenic regulation of body temperature and energy homeostasis. Endocrinology. 2010;151:4926-4937.
    • (2010) Endocrinology. , vol.151 , pp. 4926-4937
    • Roepke, T.A.1    Bosch, M.A.2    Rick, E.A.3
  • 60
    • 80053899725 scopus 로고    scopus 로고
    • Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction
    • Xu Y, Nedungadi TP, Zhu L, et al. Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metab. 2011;14:453-465.
    • (2011) Cell Metab. , vol.14 , pp. 453-465
    • Xu, Y.1    Nedungadi, T.P.2    Zhu, L.3
  • 61
    • 0032177037 scopus 로고    scopus 로고
    • Unraveling the central nervous system pathways underlying responses to leptin
    • Elmquist JK, Maratos-Flier E, Saper CB, Flier JS. Unraveling the central nervous system pathways underlying responses to leptin. Nat Neurosci. 1998;1:445-450.
    • (1998) Nat Neurosci. , vol.1 , pp. 445-450
    • Elmquist, J.K.1    Maratos-Flier, E.2    Saper, C.B.3    Flier, J.S.4
  • 62
    • 0033180532 scopus 로고    scopus 로고
    • Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area
    • Elias CF, Aschkenasi C, Lee C, et al. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron. 1999;23:775-786.
    • (1999) Neuron. , vol.23 , pp. 775-786
    • Elias, C.F.1    Aschkenasi, C.2    Lee, C.3
  • 63
    • 0033082380 scopus 로고    scopus 로고
    • From lesions to leptin: hypothalamic control of food intake and body weight
    • Elmquist JK, Elias CF, Saper CB. From lesions to leptin: hypothalamic control of food intake and body weight. Neuron. 1999;22:221-232.
    • (1999) Neuron. , vol.22 , pp. 221-232
    • Elmquist, J.K.1    Elias, C.F.2    Saper, C.B.3
  • 64
    • 0034710310 scopus 로고    scopus 로고
    • Chemical characterization of leptin-activated neurons in the rat brain
    • Elias CF, Kelly JF, Lee CE, et al. Chemical characterization of leptin-activated neurons in the rat brain. JCompNeurol. 2000;423:261-281.
    • (2000) JCompNeurol. , vol.423 , pp. 261-281
    • Elias, C.F.1    Kelly, J.F.2    Lee, C.E.3
  • 65
    • 0025166087 scopus 로고
    • Diurnal pattern of proopiomelanocortin gene expression in the arcuate nucleus of proestrous, ovariectomized, and steroid-treated rats: a possible role in cyclic luteinizing hormone secretion
    • Wise PM, Scarbrough K, Weiland NG, Larson GH. Diurnal pattern of proopiomelanocortin gene expression in the arcuate nucleus of proestrous, ovariectomized, and steroid-treated rats: a possible role in cyclic luteinizing hormone secretion. Mol Endocrinol. 1990;4:886-892.
    • (1990) Mol Endocrinol. , vol.4 , pp. 886-892
    • Wise, P.M.1    Scarbrough, K.2    Weiland, N.G.3    Larson, G.H.4
  • 66
    • 0025732991 scopus 로고
    • Changes in proopiomelanocortin messenger ribonucleic acid levels in the rostral periarcuate region of the female rat during the estrous cycle
    • Bohler HC Jr, Tracer H, Merriam GR, Petersen SL. Changes in proopiomelanocortin messenger ribonucleic acid levels in the rostral periarcuate region of the female rat during the estrous cycle. Endocrinology. 1991;128:1265-1269.
    • (1991) Endocrinology. , vol.128 , pp. 1265-1269
    • Bohler Jr., H.C.1    Tracer, H.2    Merriam, G.R.3    Petersen, S.L.4
  • 68
    • 2542561198 scopus 로고    scopus 로고
    • Expression of proopiomelanocortin and proenkephalin mRNA in sexually dimorphic brain regions are altered in adult male and female rats treated prenatally with morphine
    • Slamberová R, Hnatczuk OC, Vathy I. Expression of proopiomelanocortin and proenkephalin mRNA in sexually dimorphic brain regions are altered in adult male and female rats treated prenatally with morphine. J Pept Res. 2004;63:399-408.
    • (2004) J Pept Res. , vol.63 , pp. 399-408
    • Slamberová, R.1    Hnatczuk, O.C.2    Vathy, I.3
  • 70
    • 0036711029 scopus 로고    scopus 로고
    • Estradiol treatment fails to affect the feeding responses to melanocortin-3/4 receptor agonism or antagonism in ovariectomized rats
    • Polidori C, Geary N. Estradiol treatment fails to affect the feeding responses to melanocortin-3/4 receptor agonism or antagonism in ovariectomized rats. Peptides. 2002;23: 1697-1700.
    • (2002) Peptides. , vol.23 , pp. 1697-1700
    • Polidori, C.1    Geary, N.2
  • 72
    • 31944452253 scopus 로고    scopus 로고
    • The rise, fall, and resurrection of the ventromedial hypothalamus in the regulation of feeding behavior and body weight
    • King BM. The rise, fall, and resurrection of the ventromedial hypothalamus in the regulation of feeding behavior and body weight. Physiol Behav. 2006;87:221-244.
    • (2006) Physiol Behav. , vol.87 , pp. 221-244
    • King, B.M.1
  • 73
    • 0015908992 scopus 로고
    • Hypothalamic obesity: the myth of the ventromedial nucleus
    • Gold RM. Hypothalamic obesity: the myth of the ventromedial nucleus. Science. 1973;182:488-490.
    • (1973) Science. , vol.182 , pp. 488-490
    • Gold, R.M.1
  • 74
    • 0028959656 scopus 로고
    • The nuclear receptor steroidogenic factor 1 is essential for the formation of the ventromedial hypothalamic nucleus
    • Ikeda Y, Luo X, Abbud R, Nilson JH, Parker KL. The nuclear receptor steroidogenic factor 1 is essential for the formation of the ventromedial hypothalamic nucleus. Mol Endocrinol. 1995;9:478-486.
    • (1995) Mol Endocrinol. , vol.9 , pp. 478-486
    • Ikeda, Y.1    Luo, X.2    Abbud, R.3    Nilson, J.H.4    Parker, K.L.5
  • 75
    • 0034618213 scopus 로고    scopus 로고
    • Disruption of the gene encoding SF-1 alters the distribution of hypothalamic neuronal phenotypes
    • Dellovade TL, Young M, Ross EP, et al. Disruption of the gene encoding SF-1 alters the distribution of hypothalamic neuronal phenotypes. J Comp Neurol. 2000;423:579-589.
    • (2000) J Comp Neurol. , vol.423 , pp. 579-589
    • Dellovade, T.L.1    Young, M.2    Ross, E.P.3
  • 76
    • 0036153668 scopus 로고    scopus 로고
    • Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity
    • Majdic G, Young M, Gomez-Sanchez E, et al. Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology. 2002;143: 607-614.
    • (2002) Endocrinology. , vol.143 , pp. 607-614
    • Majdic, G.1    Young, M.2    Gomez-Sanchez, E.3
  • 77
    • 0025365407 scopus 로고
    • 17 β-Estradiol depolarization of hypothalamic neurons is mediated by cyclic AMP
    • Minami T, Oomura Y, Nabekura J, Fukuda A. 17 β-Estradiol depolarization of hypothalamic neurons is mediated by cyclic AMP. Brain Res. 1990;519:301-307.
    • (1990) Brain Res. , vol.519 , pp. 301-307
    • Minami, T.1    Oomura, Y.2    Nabekura, J.3    Fukuda, A.4
  • 78
    • 33847793319 scopus 로고    scopus 로고
    • Silencing of estrogen receptor α in the ventromedial nucleus of hypothalamus leads to metabolic syndrome
    • Musatov S, Chen W, Pfaff DW, et al. Silencing of estrogen receptor α in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. Proc Natl Acad Sci USA. 2007;104:2501-2506.
    • (2007) Proc Natl Acad Sci USA. , vol.104 , pp. 2501-2506
    • Musatov, S.1    Chen, W.2    Pfaff, D.W.3
  • 79
    • 33751175424 scopus 로고    scopus 로고
    • Sex-specific densities of estrogen receptorsα andβ in the subnuclei of the nucleus tractus solitarius, hypoglossal nucleus and dorsal vagal motor nucleus weanling rats
    • Schlenker EH, Hansen SN. Sex-specific densities of estrogen receptorsα andβ in the subnuclei of the nucleus tractus solitarius, hypoglossal nucleus and dorsal vagal motor nucleus weanling rats. Brain Res. 2006;1123:89-100.
    • (2006) Brain Res. , vol.1123 , pp. 89-100
    • Schlenker, E.H.1    Hansen, S.N.2
  • 80
    • 36349004794 scopus 로고    scopus 로고
    • Estradiol enhances cholecystokinindependent lipid-induced satiation and activates estrogen receptor-α-expressing cells in the nucleus tractus solitarius of ovariectomized rats
    • Asarian L, Geary N. Estradiol enhances cholecystokinindependent lipid-induced satiation and activates estrogen receptor-α-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology. 2007;148:5656-5666.
    • (2007) Endocrinology. , vol.148 , pp. 5656-5666
    • Asarian, L.1    Geary, N.2
  • 81
    • 0034741811 scopus 로고    scopus 로고
    • Estradiol, CCK and satiation
    • Geary N. Estradiol, CCK and satiation. Peptides. 2001; 22:1251-1263.
    • (2001) Peptides. , vol.22 , pp. 1251-1263
    • Geary, N.1
  • 82
    • 64949123113 scopus 로고    scopus 로고
    • Gut peptides in the control of food intake
    • Moran TH. Gut peptides in the control of food intake. Int J Obes (Lond). 2009;33(suppl 1):S7-S10.
    • (2009) Int J Obes (Lond). , vol.33 , Issue.SUPPL. 1
    • Moran, T.H.1
  • 83
    • 0034910191 scopus 로고    scopus 로고
    • The effect of cholecystokinin in controlling appetite and food intake in humans
    • Degen L, Matzinger D, Drewe J, Beglinger C. The effect of cholecystokinin in controlling appetite and food intake in humans. Peptides. 2001;22:1265-1269.
    • (2001) Peptides. , vol.22 , pp. 1265-1269
    • Degen, L.1    Matzinger, D.2    Drewe, J.3    Beglinger, C.4
  • 84
    • 0029934805 scopus 로고    scopus 로고
    • The increased satiating potency of CCK-8 by estradiol is not mediated by upregulation of NTS CCK receptors
    • Geary N, Smith GP, Corp ES. The increased satiating potency of CCK-8 by estradiol is not mediated by upregulation of NTS CCK receptors. Brain Res. 1996;719: 179-186.
    • (1996) Brain Res. , vol.719 , pp. 179-186
    • Geary, N.1    Smith, G.P.2    Corp, E.S.3
  • 86
    • 0027156856 scopus 로고
    • Modulation of the satiety effect of cholecystokinin by estradiol
    • Butera PC, Bradway DM, Cataldo NJ. Modulation of the satiety effect of cholecystokinin by estradiol. Physiol Behav. 1993;53:1235-1238.
    • (1993) Physiol Behav. , vol.53 , pp. 1235-1238
    • Butera, P.C.1    Bradway, D.M.2    Cataldo, N.J.3
  • 87
    • 0032888418 scopus 로고    scopus 로고
    • Cyclic estradiol treatment phasically potentiates endogenous cholecystokinin's satiating action in ovariectomized rats
    • Asarian L, Geary N. Cyclic estradiol treatment phasically potentiates endogenous cholecystokinin's satiating action in ovariectomized rats. Peptides. 1999;20:445-450.
    • (1999) Peptides. , vol.20 , pp. 445-450
    • Asarian, L.1    Geary, N.2
  • 88
    • 41549130400 scopus 로고    scopus 로고
    • Hindbrain administration of estradiol inhibits feeding and activates estrogen receptor-α-expressing cells in the nucleus tractus solitarius of ovariectomized rats
    • Thammacharoen S, Lutz TA, Geary N, Asarian L. Hindbrain administration of estradiol inhibits feeding and activates estrogen receptor-α-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology. 2008;149:1609-1617.
    • (2008) Endocrinology. , vol.149 , pp. 1609-1617
    • Thammacharoen, S.1    Lutz, T.A.2    Geary, N.3    Asarian, L.4
  • 92
    • 0029908224 scopus 로고    scopus 로고
    • Central infusion of leptin and GLP-1 (7-36) amide differentially stimulate c-Fos-like immunoreactivity in the rat brain
    • van Dijk G, Thiele TE, Donahey JC, et al. Central infusion of leptin and GLP-1 (7-36) amide differentially stimulate c-Fos-like immunoreactivity in the rat brain.AmJ Physiol. 1996;271:R1096-R1100.
    • (1996) AmJ Physiol. , vol.271
    • van Dijk, G.1    Thiele, T.E.2    Donahey, J.C.3
  • 93
    • 0031906674 scopus 로고    scopus 로고
    • Leptin activates distinct projections from the dorsomedial and ventromedial hypothalamic nuclei
    • Elmquist JK, Ahima RS, Elias CF, Flier JS, Saper CB. Leptin activates distinct projections from the dorsomedial and ventromedial hypothalamic nuclei. Proc Natl Acad Sci USA. 1998;95:741-746.
    • (1998) Proc Natl Acad Sci USA. , vol.95 , pp. 741-746
    • Elmquist, J.K.1    Ahima, R.S.2    Elias, C.F.3    Flier, J.S.4    Saper, C.B.5
  • 95
    • 0032561726 scopus 로고    scopus 로고
    • Leptin receptors in estrogen receptor-containing neurons of the female rat hypothalamus
    • 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.
    • (1998) Brain Res. , vol.812 , pp. 256-259
    • Diano, S.1    Kalra, S.P.2    Sakamoto, H.3    Horvath, T.L.4
  • 96
    • 0033047926 scopus 로고    scopus 로고
    • Cyclical variations in the abundance of leptin receptors, but not in circulating leptin, correlate with NPY expression during the oestrous cycle
    • Bennett PA, Lindell K, Wilson C, CarlssonLM,Carlsson B, Robinson IC. Cyclical variations in the abundance of leptin receptors, but not in circulating leptin, correlate with NPY expression during the oestrous cycle. Neuroendocrinology. 1999;69:417-423.
    • (1999) Neuroendocrinology. , vol.69 , pp. 417-423
    • Bennett, P.A.1    Lindell, K.2    Wilson, C.3    Carlsson, L.M.4    Carlsson, B.5    Robinson, I.C.6
  • 97
    • 0035857472 scopus 로고    scopus 로고
    • Leptin receptor 5βuntranslated regions in the rat: relative abundance, genomic organization and relation to putative response elements
    • Lindell K, Bennett PA, Itoh Y, Robinson IC, Carlsson LM, Carlsson B. Leptin receptor 5βuntranslated regions in the rat: relative abundance, genomic organization and relation to putative response elements. Mol Cell Endocrinol. 2001; 172:37-45.
    • (2001) Mol Cell Endocrinol. , vol.172 , pp. 37-45
    • Lindell, K.1    Bennett, P.A.2    Itoh, Y.3    Robinson, I.C.4    Carlsson, L.M.5    Carlsson, B.6
  • 98
    • 0035163612 scopus 로고    scopus 로고
    • Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y
    • Ainslie DA, Morris MJ, Wittert G, Turnbull H, Proietto J, Thorburn AW. Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y. Int J Obes Relat Metab Disord. 2001;25:1680-1688. 99. Clegg DJ, Riedy CA, Smith KA, Benoit SC, Woods SC. Differential sensitivity to central leptin and insulin in male and female rats. Diabetes. 2003;52:682-687.
    • (2001) Int J Obes Relat Metab Disord. , vol.25 , pp. 1680-1688
    • Ainslie, D.A.1    Morris, M.J.2    Wittert, G.3    Turnbull, H.4    Proietto, J.5    Thorburn, A.W.6
  • 99
    • 0344406153 scopus 로고    scopus 로고
    • Differential sensitivity to central leptin and insulin in male and female rats
    • Clegg DJ, Riedy CA, Smith KA, Benoit SC, Woods SC. Differential sensitivity to central leptin and insulin in male and female rats. Diabetes. 2003;52:682-687.
    • (2003) Diabetes. , vol.52 , pp. 682-687
    • Clegg, D.J.1    Riedy, C.A.2    Smith, K.A.3    Benoit, S.C.4    Woods, S.C.5
  • 100
    • 33745298324 scopus 로고    scopus 로고
    • Gonadal hormones determine sensitivity to central leptin and insulin
    • Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55:978-987.
    • (2006) Diabetes. , vol.55 , pp. 978-987
    • Clegg, D.J.1    Brown, L.M.2    Woods, S.C.3    Benoit, S.C.4
  • 101
    • 4243761409 scopus 로고
    • 1995 ventricular insulin infusion attenuates NPY-induced feeding at the level of the paraventricular nucleus
    • Chavez M, van Dijk G, Arkies BJ, Woods SC. 1995 ventricular insulin infusion attenuates NPY-induced feeding at the level of the paraventricular nucleus. Obes Res. 1995; 3:335s
    • (1995) Obes Res. , vol.3
    • Chavez, M.1    van Dijk, G.2    Arkies, B.J.3    Woods, S.C.4
  • 102
    • 0032980181 scopus 로고    scopus 로고
    • ArcuateNPYneurons and energy homeostasis in diet-induced obese and resistant rats
    • Levin BE. ArcuateNPYneurons and energy homeostasis in diet-induced obese and resistant rats. Am J Physiol. 1999; 276:R382-R387.
    • (1999) Am J Physiol. , vol.276
    • Levin, B.E.1
  • 104
    • 0242667851 scopus 로고    scopus 로고
    • Neuropeptide Y and energy homeostasis: insights fromYreceptor knockout models
    • Herzog H. Neuropeptide Y and energy homeostasis: insights fromYreceptor knockout models. Eur J Pharmacol. 2003;480:21-29.
    • (2003) Eur J Pharmacol. , vol.480 , pp. 21-29
    • Herzog, H.1
  • 105
    • 0033065587 scopus 로고    scopus 로고
    • Leptin receptor long-form splice-variant protein expression in neuron cell bodies of the brain and colocalization with neuropeptideY mRNA in the arcuate nucleus
    • Baskin DG, SchwartzMW,Seeley RJ, et al. Leptin receptor long-form splice-variant protein expression in neuron cell bodies of the brain and colocalization with neuropeptideY mRNA in the arcuate nucleus. J Histochem Cytochem. 1999;47:353-362.
    • (1999) J Histochem Cytochem. , vol.47 , pp. 353-362
    • Baskin, D.G.1    Schwartz, M.W.2    Seeley, R.J.3
  • 106
    • 2042481350 scopus 로고    scopus 로고
    • Estrogen induces neuropeptide Y (NPY) Y1 receptor gene expression and responsiveness to NPY in gonadotrope-enriched pituitary cell cultures
    • Hill JW, Urban JH, Xu M, Levine JE. Estrogen induces neuropeptide Y (NPY) Y1 receptor gene expression and responsiveness to NPY in gonadotrope-enriched pituitary cell cultures. Endocrinology. 2004;145:2283-2290.
    • (2004) Endocrinology. , vol.145 , pp. 2283-2290
    • Hill, J.W.1    Urban, J.H.2    Xu, M.3    Levine, J.E.4
  • 107
    • 0028318098 scopus 로고
    • Anorectic effects of estrogen may be mediated by decreased neuropeptide-Y release in the hypothalamic paraventricular nucleus
    • Bonavera JJ, Dube MG, Kalra PS, Kalra SP. Anorectic effects of estrogen may be mediated by decreased neuropeptide-Y release in the hypothalamic paraventricular nucleus. Endocrinology. 1994;134:2367-2370.
    • (1994) Endocrinology. , vol.134 , pp. 2367-2370
    • Bonavera, J.J.1    Dube, M.G.2    Kalra, P.S.3    Kalra, S.P.4
  • 108
    • 33747813597 scopus 로고    scopus 로고
    • Coordinate regulation of neuropeptideYand agouti-related peptide gene expression by estrogen depends on the ratio of estrogen receptor (ER) α toERβ in clonal hypothalamic neurons
    • Titolo D, Cai F, Belsham DD. Coordinate regulation of neuropeptideYand agouti-related peptide gene expression by estrogen depends on the ratio of estrogen receptor (ER) α toERβ in clonal hypothalamic neurons.MolEndocrinol. 2006;20:2080-2092.
    • (2006) MolEndocrinol. , vol.20 , pp. 2080-2092
    • Titolo, D.1    Cai, F.2    Belsham, D.D.3
  • 109
    • 70349452304 scopus 로고    scopus 로고
    • Functional requirement of AgRP andNPYneurons in ovarian cycle-dependent regulation of food intake
    • Olofsson LE, Pierce AA, Xu AW. Functional requirement of AgRP andNPYneurons in ovarian cycle-dependent regulation of food intake. Proc Natl Acad Sci USA. 2009;106: 15932-15937.
    • (2009) Proc Natl Acad Sci USA. , vol.106 , pp. 15932-15937
    • Olofsson, L.E.1    Pierce, A.A.2    Xu, A.W.3
  • 110
    • 0027400507 scopus 로고
    • Genetic and nongenetic determinants of regional fat distribution
    • Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocr Rev. 1993;14:72-93.
    • (1993) Endocr Rev. , vol.14 , pp. 72-93
    • Bouchard, C.1    Despres, J.P.2    Mauriege, P.3
  • 111
    • 0023035033 scopus 로고
    • Subcutaneous and visceral fat distribution according to sex, age, and overweight, evaluated by computed tomography
    • Enzi G, Gasparo M, Biondetti PR, Fiore D, Semisa M, Zurlo F. Subcutaneous and visceral fat distribution according to sex, age, and overweight, evaluated by computed tomography. Am J Clin Nutr. 1986;44:739-746.
    • (1986) Am J Clin Nutr. , vol.44 , pp. 739-746
    • Enzi, G.1    Gasparo, M.2    Biondetti, P.R.3    Fiore, D.4    Semisa, M.5    Zurlo, F.6
  • 112
    • 0026538546 scopus 로고
    • Abdominal fat distribution and disease: an overview of epidemiological data
    • Bjorntorp P. Abdominal fat distribution and disease: an overview of epidemiological data. Ann Med. 1992;24: 15-18. 113. Bjorntorp P. Abdominal fat distribution and the metabolic syndrome. J Cardiovasc Pharmacol. 1992;20(suppl 8): S26-S28.
    • (1992) Ann Med. , vol.24 , pp. 15-18
    • Bjorntorp, P.1
  • 113
    • 0027062816 scopus 로고
    • Abdominal fat distribution and the metabolic syndrome
    • Bjorntorp P. Abdominal fat distribution and the metabolic syndrome. J Cardiovasc Pharmacol. 1992;20(suppl 8): S26-S28.
    • (1992) J Cardiovasc Pharmacol. , vol.20 , Issue.SUPPL. 8
    • Bjorntorp, P.1
  • 114
    • 0026727710 scopus 로고    scopus 로고
    • 1992 Hormonal effects on fat distribution and its relationship to health risk factors
    • discussion 61
    • Bjørntorp P. 1992 Hormonal effects on fat distribution and its relationship to health risk factors. Acta Paediatr Suppl 383:59-60; discussion 61.
    • Acta Paediatr Suppl , vol.383 , pp. 59-60
    • Bjørntorp, P.1
  • 115
    • 10944236220 scopus 로고    scopus 로고
    • The menopausal transition a 9-year prospective population-based study The MelbourneWomen'sMidlife Health Project
    • Guthrie JR, Dennerstein L, Taffe JR, Lehert P, Burger HG. The menopausal transition a 9-year prospective population-based study The MelbourneWomen'sMidlife Health Project. Climacteric. 2004;7:375-389.
    • (2004) Climacteric. , vol.7 , pp. 375-389
    • Guthrie, J.R.1    Dennerstein, L.2    Taffe, J.R.3    Lehert, P.4    Burger, H.G.5
  • 116
    • 47249146881 scopus 로고    scopus 로고
    • Increased visceral fat and decreased energy expenditure during the menopausal transition
    • Lovejoy JC, ChampagneCM,de Jonge L, Xie H, Smith SR. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes (Lond). 2008;32:949-958.
    • (2008) Int J Obes (Lond). , vol.32 , pp. 949-958
    • Lovejoy, J.C.1    Champagne, C.M.2    de Jonge, L.3    Xie, H.4    Smith, S.R.5
  • 117
    • 0031032354 scopus 로고    scopus 로고
    • Body weight, body fat distribution, and hormonal replacement therapy in early postmenopausal women
    • Gambacciani M, Ciaponi M, Cappagli B, et al. Body weight, body fat distribution, and hormonal replacement therapy in early postmenopausal women. J Clin Endocrinol Metab. 1997;82:414-417.
    • (1997) J Clin Endocrinol Metab. , vol.82 , pp. 414-417
    • Gambacciani, M.1    Ciaponi, M.2    Cappagli, B.3
  • 118
    • 0025910420 scopus 로고
    • Hormone replacement therapy prevents coronary artery disease in ovariectomized cholesterol-fed rabbits
    • Haarbo J, Hansen BF, Christiansen C. Hormone replacement therapy prevents coronary artery disease in ovariectomized cholesterol-fed rabbits. APMIS. 1991;99:721-727.
    • (1991) APMIS. , vol.99 , pp. 721-727
    • Haarbo, J.1    Hansen, B.F.2    Christiansen, C.3
  • 119
    • 0026347185 scopus 로고
    • Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause
    • Haarbo J, Marslew U, Gotfredsen A, Christiansen C. Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause. Metabolism. 1991;40:1323-1326.
    • (1991) Metabolism. , vol.40 , pp. 1323-1326
    • Haarbo, J.1    Marslew, U.2    Gotfredsen, A.3    Christiansen, C.4
  • 120
    • 0032986811 scopus 로고    scopus 로고
    • Effects of sex steroid hormones on regional fat depots as assessed by magnetic resonance imaging in transsexuals
    • Elbers JM, Asscheman H, Seidell JC, Gooren LJ. Effects of sex steroid hormones on regional fat depots as assessed by magnetic resonance imaging in transsexuals.AmJ Physiol. 1999;276:E317-E325.
    • (1999) AmJ Physiol. , vol.276
    • Elbers, J.M.1    Asscheman, H.2    Seidell, J.C.3    Gooren, L.J.4
  • 121
    • 0034504237 scopus 로고    scopus 로고
    • Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome
    • Wajchenberg BL. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr Rev. 2000;21:697-738.
    • (2000) Endocr Rev. , vol.21 , pp. 697-738
    • Wajchenberg, B.L.1
  • 122
    • 0036179818 scopus 로고    scopus 로고
    • A pilot study of long-term effects of a novel obesity treatment: omentectomy in connection with adjustable gastric banding
    • Thorne A, Lonnqvist F, Apelman J, Hellers G, Arner P. A pilot study of long-term effects of a novel obesity treatment: omentectomy in connection with adjustable gastric banding. Int J Obes Relat Metab Disord. 2002;26:193-199.
    • (2002) Int J Obes Relat Metab Disord. , vol.26 , pp. 193-199
    • Thorne, A.1    Lonnqvist, F.2    Apelman, J.3    Hellers, G.4    Arner, P.5
  • 123
    • 77958176309 scopus 로고    scopus 로고
    • Potential additional effect of omentectomy on metabolic syndrome, acute-phase reactants, and inflammatory mediators in grade III obese patients undergoing laparoscopic Roux-en-Y gastric bypass: a randomized trial
    • Herrera MF, Pantoja JP, Velazquez-Fernandez D, et al. Potential additional effect of omentectomy on metabolic syndrome, acute-phase reactants, and inflammatory mediators in grade III obese patients undergoing laparoscopic Roux-en-Y gastric bypass: a randomized trial. Diabetes Care. 2010;33:1413-1418.
    • (2010) Diabetes Care. , vol.33 , pp. 1413-1418
    • Herrera, M.F.1    Pantoja, J.P.2    Velazquez-Fernandez, D.3
  • 124
    • 77955485237 scopus 로고    scopus 로고
    • Surgical removal of omental fat does not improve insulin sensitivity and cardiovascular risk factors in obese adults
    • Fabbrini E, Tamboli RA, Magkos F, et al. Surgical removal of omental fat does not improve insulin sensitivity and cardiovascular risk factors in obese adults. Gastroenterology. 2010;139:448-455.
    • (2010) Gastroenterology. , vol.139 , pp. 448-455
    • Fabbrini, E.1    Tamboli, R.A.2    Magkos, F.3
  • 125
    • 0036787345 scopus 로고    scopus 로고
    • Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process?
    • Gabriely I, Ma XH, Yang XM, et al. Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process? Diabetes. 2002;51: 2951-2958.
    • (2002) Diabetes , vol.51 , pp. 2951-2958
    • Gabriely, I.1    Ma, X.H.2    Yang, X.M.3
  • 126
    • 0021678265 scopus 로고
    • Distribution of adipose tissue and risk of cardiovascular disease and death: a 12 year follow up of participants in the population study of women in Gothenburg, Sweden
    • Lapidus L, Bengtsson C, Larsson B, Pennert K, Rybo E, Sjostrom L. Distribution of adipose tissue and risk of cardiovascular disease and death: a 12 year follow up of participants in the population study of women in Gothenburg, Sweden. Br Med J (Clin Res Ed). 1984;289:1257-1261.
    • (1984) Br Med J (Clin Res Ed). , vol.289 , pp. 1257-1261
    • Lapidus, L.1    Bengtsson, C.2    Larsson, B.3    Pennert, K.4    Rybo, E.5    Sjostrom, L.6
  • 128
    • 3042803327 scopus 로고
    • Central obesity and coronary heart disease in men
    • Donahue RP, Abbott RD. Central obesity and coronary heart disease in men. Lancet. 1987;2:1215.
    • (1987) Lancet. , vol.2 , pp. 1215
    • Donahue, R.P.1    Abbott, R.D.2
  • 129
    • 0022408652 scopus 로고
    • The influence of body fat distribution on the incidence of diabetes mellitus: 13.5 years of follow-up of the participants in the study of men born in 1913
    • Ohlson LO, Larsson B, Svardsudd K, et al. The influence of body fat distribution on the incidence of diabetes mellitus: 13.5 years of follow-up of the participants in the study of men born in 1913. Diabetes. 1985;34:1055-1058.
    • (1985) Diabetes. , vol.34 , pp. 1055-1058
    • Ohlson, L.O.1    Larsson, B.2    Svardsudd, K.3
  • 130
    • 0029671107 scopus 로고    scopus 로고
    • The android woman-a risky condition
    • Bjorntorp P. The android woman-a risky condition. J Intern Med. 1996;239:105-110.
    • (1996) J Intern Med. , vol.239 , pp. 105-110
    • Bjorntorp, P.1
  • 131
    • 42649144463 scopus 로고    scopus 로고
    • Beneficial ef-fects of subcutaneous fat transplantation on metabolism
    • Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial ef-fects of subcutaneous fat transplantation on metabolism. Cell Metab. 2008;7:410-420.
    • (2008) Cell Metab. , vol.7 , pp. 410-420
    • Tran, T.T.1    Yamamoto, Y.2    Gesta, S.3    Kahn, C.R.4
  • 134
    • 84995853519 scopus 로고
    • Identification of estrogen receptor in human adipose tissue and adipocytes
    • Mizutani T, Nishikawa Y, Adachi H, et al. Identification of estrogen receptor in human adipose tissue and adipocytes. J Clin Endocrinol Metab. 1994;78:950-954.
    • (1994) J Clin Endocrinol Metab. , vol.78 , pp. 950-954
    • Mizutani, T.1    Nishikawa, Y.2    Adachi, H.3
  • 135
    • 0027379181 scopus 로고
    • Determination of estrogen receptor messenger ribonucleic acid (mRNA) and cytochrome P450 aromatasemRNAlevels in adipocytes and adipose stromal cells by competitive polymerase chain reaction amplification
    • PriceTM,O'Brien SN. Determination of estrogen receptor messenger ribonucleic acid (mRNA) and cytochrome P450 aromatasemRNAlevels in adipocytes and adipose stromal cells by competitive polymerase chain reaction amplification. J Clin Endocrinol Metab. 1993;77:1041-1045.
    • (1993) J Clin Endocrinol Metab. , vol.77 , pp. 1041-1045
    • Price, T.M.1    O'Brien, S.N.2
  • 136
    • 0033607207 scopus 로고    scopus 로고
    • Survival of reproductive behaviors in estrogen receptor β gene-deficient (βERKO) male and female mice
    • Ogawa S, Chan J, Chester AE, Gustafsson JA, Korach KS, Pfaff DW. Survival of reproductive behaviors in estrogen receptor β gene-deficient (βERKO) male and female mice. Proc Natl Acad Sci USA. 1999;96:12887-12892.
    • (1999) Proc Natl Acad Sci USA. , vol.96 , pp. 12887-12892
    • Ogawa, S.1    Chan, J.2    Chester, A.E.3    Gustafsson, J.A.4    Korach, K.S.5    Pfaff, D.W.6
  • 137
    • 0028143234 scopus 로고
    • Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man
    • Smith EP, Boyd J, Frank GR, et al. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med. 1994;331:1056-1061.
    • (1994) N Engl J Med. , vol.331 , pp. 1056-1061
    • Smith, E.P.1    Boyd, J.2    Frank, G.R.3
  • 139
    • 34249659045 scopus 로고    scopus 로고
    • Oestrogen receptor α gene expression levels are reduced in obese compared to normal weight females
    • Nilsson M, Dahlman I, Ryden M, et al. Oestrogen receptor α gene expression levels are reduced in obese compared to normal weight females. Int J Obes (Lond). 2007;31:900-907.
    • (2007) Int J Obes (Lond). , vol.31 , pp. 900-907
    • Nilsson, M.1    Dahlman, I.2    Ryden, M.3
  • 140
    • 0034458392 scopus 로고    scopus 로고
    • Association of estrogen receptor-α genotypes with body mass index in normal healthy postmenopausal Caucasian women
    • Deng HW, Li J, Li JL, et al. Association of estrogen receptor-α genotypes with body mass index in normal healthy postmenopausal Caucasian women. J Clin Endocrinol Metab. 2000;85:2748-2751.
    • (2000) J Clin Endocrinol Metab. , vol.85 , pp. 2748-2751
    • Deng, H.W.1    Li, J.2    Li, J.L.3
  • 141
    • 77955565430 scopus 로고    scopus 로고
    • The association between the rs2234693 and rs9340799 estrogen receptor α gene polymorphisms and risk factors for cardiovascular disease: a review
    • Casazza K, Page GP, Fernandez JR. The association between the rs2234693 and rs9340799 estrogen receptor α gene polymorphisms and risk factors for cardiovascular disease: a review. Biol Res Nurs. 2010;12:84-97.
    • (2010) Biol Res Nurs. , vol.12 , pp. 84-97
    • Casazza, K.1    Page, G.P.2    Fernandez, J.R.3
  • 142
    • 0036019521 scopus 로고    scopus 로고
    • Association of polymorphisms of the estrogen receptorα gene with bone mineral density of the femoral neck in elderly Japanese women
    • Yamada Y, Ando F, Niino N, Ohta S, Shimokata H. Association of polymorphisms of the estrogen receptorα gene with bone mineral density of the femoral neck in elderly Japanese women. J Mol Med. 2002;80:452-460.
    • (2002) J Mol Med. , vol.80 , pp. 452-460
    • Yamada, Y.1    Ando, F.2    Niino, N.3    Ohta, S.4    Shimokata, H.5
  • 143
    • 0037271644 scopus 로고    scopus 로고
    • Relationships of resting energy expenditure with body fat distribution and abdominal fatness in Japanese population
    • Okura T, Koda M, Ando F, Niino N, Shimokata H. Relationships of resting energy expenditure with body fat distribution and abdominal fatness in Japanese population. J Physiol Anthropol Appl Human Sci. 2003;22: 47-52.
    • (2003) J Physiol Anthropol Appl Human Sci. , vol.22 , pp. 47-52
    • Okura, T.1    Koda, M.2    Ando, F.3    Niino, N.4    Shimokata, H.5
  • 144
    • 0141920798 scopus 로고    scopus 로고
    • Association of the mitochondrial DNA 15497G/A polymorphism with obesity in a middle-aged and elderly Japanese population
    • Okura T, Koda M, Ando F, Niino N, Tanaka M, Shimokata H. Association of the mitochondrial DNA 15497G/A polymorphism with obesity in a middle-aged and elderly Japanese population. Hum Genet. 2003;113: 432-436.
    • (2003) Hum Genet. , vol.113 , pp. 432-436
    • Okura, T.1    Koda, M.2    Ando, F.3    Niino, N.4    Tanaka, M.5    Shimokata, H.6
  • 148
    • 56049110515 scopus 로고    scopus 로고
    • Mechanisms of antidiabetogenic and body weight-lowering effects of estrogen in high-fat diet-fed mice
    • Bryzgalova G, Lundholm L, Portwood N, et al. Mechanisms of antidiabetogenic and body weight-lowering effects of estrogen in high-fat diet-fed mice. Am J Physiol Endocrinol Metab. 2008;295:E904-E912.
    • (2008) Am J Physiol Endocrinol Metab. , vol.295
    • Bryzgalova, G.1    Lundholm, L.2    Portwood, N.3
  • 149
    • 33744492590 scopus 로고    scopus 로고
    • Long-term administration of estradiol decreases expression of hepatic lipogenic genes and improves insulin sensitivity in ob/ob mice: a possible mechanism is through direct regulation of signal transducer and activator of transcription 3
    • Gao H, Bryzgalova G, Hedman E, et al. Long-term administration of estradiol decreases expression of hepatic lipogenic genes and improves insulin sensitivity in ob/ob mice: a possible mechanism is through direct regulation of signal transducer and activator of transcription 3. Mol Endocrinol. 2006;20:1287-1299.
    • (2006) Mol Endocrinol. , vol.20 , pp. 1287-1299
    • Gao, H.1    Bryzgalova, G.2    Hedman, E.3
  • 150
    • 56849109099 scopus 로고    scopus 로고
    • The estrogen receptor α-selective agonist propyl pyrazole triol improves glucose tolerance in ob/ob mice; potential molecular mechanisms
    • Lundholm L, Bryzgalova G, Gao H, et al. The estrogen receptor α-selective agonist propyl pyrazole triol improves glucose tolerance in ob/ob mice; potential molecular mechanisms. J Endocrinol. 2008;199:275-286.
    • (2008) J Endocrinol. , vol.199 , pp. 275-286
    • Lundholm, L.1    Bryzgalova, G.2    Gao, H.3
  • 151
    • 0024386369 scopus 로고
    • Insulin regulation of lipoprotein lipase activity in 3T3-L1 adipocytes is mediated at posttranscriptional and posttranslational levels
    • Semenkovich CF, Wims M, Noe L, Etienne J, Chan L. Insulin regulation of lipoprotein lipase activity in 3T3-L1 adipocytes is mediated at posttranscriptional and posttranslational levels. J Biol Chem. 1989;264:9030-9038.
    • (1989) J Biol Chem. , vol.264 , pp. 9030-9038
    • Semenkovich, C.F.1    Wims, M.2    Noe, L.3    Etienne, J.4    Chan, L.5
  • 152
    • 0023778715 scopus 로고
    • Relationship between lipoprotein lipase activity and plasma sex steroid level in obese women
    • Iverius PH, Brunzell JD. Relationship between lipoprotein lipase activity and plasma sex steroid level in obese women. J Clin Invest. 1988;82:1106-1112.
    • (1988) J Clin Invest. , vol.82 , pp. 1106-1112
    • Iverius, P.H.1    Brunzell, J.D.2
  • 153
    • 0030334820 scopus 로고    scopus 로고
    • Effect of estrogen replacement therapy on hepatic triglyceride lipase, lipoprotein lipase and lipids including apolipoprotein E in climacteric and elderly women
    • Urabe M, Yamamoto T, Kashiwagi T, et al. Effect of estrogen replacement therapy on hepatic triglyceride lipase, lipoprotein lipase and lipids including apolipoprotein E in climacteric and elderly women. Endocr J. 1996;43:737-742.
    • (1996) Endocr J. , vol.43 , pp. 737-742
    • Urabe, M.1    Yamamoto, T.2    Kashiwagi, T.3
  • 155
    • 84863494132 scopus 로고    scopus 로고
    • Lipin proteins and metabolic homeostasis
    • Reue K, Dwyer JR. Lipin proteins and metabolic homeostasis. J Lipid Res. 2009;50 suppl:S109-S114.
    • (2009) J Lipid Res. , vol.50 , Issue.SUPPL.
    • Reue, K.1    Dwyer, J.R.2
  • 156
    • 35348903414 scopus 로고    scopus 로고
    • Human subcutaneous adipose tissue LPIN1 expression in obesity, type 2 diabetes mellitus, and human immunodeficiency virus-associated lipodystrophy syndrome
    • Miranda M, Chacon MR, Gomez J, et al. Human subcutaneous adipose tissue LPIN1 expression in obesity, type 2 diabetes mellitus, and human immunodeficiency virus-associated lipodystrophy syndrome. Metabolism. 2007; 56:1518-1526.
    • (2007) Metabolism. , vol.56 , pp. 1518-1526
    • Miranda, M.1    Chacon, M.R.2    Gomez, J.3
  • 157
    • 33845980542 scopus 로고    scopus 로고
    • A role of lipin in human obesity and insulin resistance: relation to adipocyte glucose transport andGLUT4expression
    • van Harmelen V, Ryden M, Sjolin E, Hoffstedt J. A role of lipin in human obesity and insulin resistance: relation to adipocyte glucose transport andGLUT4expression. J Lipid Res. 2007;48:201-206.
    • (2007) J Lipid Res. , vol.48 , pp. 201-206
    • van Harmelen, V.1    Ryden, M.2    Sjolin, E.3    Hoffstedt, J.4
  • 158
    • 80855132321 scopus 로고    scopus 로고
    • Regulation of lipin1 by nutritional status, adiponectin, sex and pituitary function in rat white adipose tissue
    • Gonzalez CR, Novelle MG, Caminos JE, et al. Regulation of lipin1 by nutritional status, adiponectin, sex and pituitary function in rat white adipose tissue. Physiol Behav. 2012;105:777-783.
    • (2012) Physiol Behav. , vol.105 , pp. 777-783
    • Gonzalez, C.R.1    Novelle, M.G.2    Caminos, J.E.3
  • 159
    • 10044259640 scopus 로고    scopus 로고
    • Role of estrogens in adipocyte development and function
    • Cooke PS, Naaz A. Role of estrogens in adipocyte development and function. Exp Biol Med (Maywood). 2004; 229:1127-1135.
    • (2004) Exp Biol Med (Maywood). , vol.229 , pp. 1127-1135
    • Cooke, P.S.1    Naaz, A.2
  • 160
    • 46249083607 scopus 로고    scopus 로고
    • Metabolic actions of estrogen receptor β (ERβ) are mediated by a negative cross-talk with PPARγ
    • Foryst-Ludwig A, Clemenz M, Hohmann S, et al. Metabolic actions of estrogen receptor β (ERβ) are mediated by a negative cross-talk with PPARγ. PLoS Genet. 2008;4: e1000108.
    • (2008) PLoS Genet. , vol.4
    • Foryst-Ludwig, A.1    Clemenz, M.2    Hohmann, S.3
  • 161
    • 0037463608 scopus 로고    scopus 로고
    • The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988-1994
    • Park YW, Zhu S, Palaniappan L, Heshka S, Carnethon MR, Heymsfield SB. The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988-1994. Arch Intern Med. 2003;163: 427-436.
    • (2003) Arch Intern Med. , vol.163 , pp. 427-436
    • Park, Y.W.1    Zhu, S.2    Palaniappan, L.3    Heshka, S.4    Carnethon, M.R.5    Heymsfield, S.B.6
  • 162
    • 0021723041 scopus 로고
    • Sex and insulin sensitivity
    • Yki-Jarvinen H. Sex and insulin sensitivity. Metabolism. 1984;33:1011-1015.
    • (1984) Metabolism. , vol.33 , pp. 1011-1015
    • Yki-Jarvinen, H.1
  • 163
    • 34249907880 scopus 로고    scopus 로고
    • Macrophage PPAR γ is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones
    • Hevener AL, Olefsky JM, Reichart D, et al. Macrophage PPAR γ is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones. J Clin Invest. 2007;117:1658-1669.
    • (2007) J Clin Invest. , vol.117 , pp. 1658-1669
    • Hevener, A.L.1    Olefsky, J.M.2    Reichart, D.3
  • 164
    • 34447130778 scopus 로고    scopus 로고
    • Overexpression of uncoupling protein 3 in skeletal muscle protects against fat-induced insulin resistance
    • Choi CS, Fillmore JJ, Kim JK, et al. Overexpression of uncoupling protein 3 in skeletal muscle protects against fat-induced insulin resistance. J Clin Invest. 2007;117: 1995-2003.
    • (2007) J Clin Invest. , vol.117 , pp. 1995-2003
    • Choi, C.S.1    Fillmore, J.J.2    Kim, J.K.3
  • 165
    • 0034982369 scopus 로고    scopus 로고
    • Decreased susceptibility to fatty acidinduced peripheral tissue insulin resistance in women
    • Frias JP, Macaraeg GB, Ofrecio J, Yu JG, Olefsky JM, Kruszynska YT. Decreased susceptibility to fatty acidinduced peripheral tissue insulin resistance in women. Diabetes. 2001;50:1344-1350.
    • (2001) Diabetes. , vol.50 , pp. 1344-1350
    • Frias, J.P.1    Macaraeg, G.B.2    Ofrecio, J.3    Yu, J.G.4    Olefsky, J.M.5    Kruszynska, Y.T.6
  • 166
    • 0036266899 scopus 로고    scopus 로고
    • Female rats do not exhibit free fatty acid-induced insulin resistance
    • Hevener A, Reichart D, Janez A, Olefsky J. Female rats do not exhibit free fatty acid-induced insulin resistance. Diabetes. 2002;51:1907-1912.
    • (2002) Diabetes. , vol.51 , pp. 1907-1912
    • Hevener, A.1    Reichart, D.2    Janez, A.3    Olefsky, J.4
  • 167
    • 0032530765 scopus 로고    scopus 로고
    • A genderrelated defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor α-deficient mice
    • Djouadi F, Weinheimer CJ, Saffitz JE, et al. A genderrelated defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor α-deficient mice. J Clin Invest. 1998;102:1083-1091.
    • (1998) J Clin Invest. , vol.102 , pp. 1083-1091
    • Djouadi, F.1    Weinheimer, C.J.2    Saffitz, J.E.3
  • 168
    • 61849113209 scopus 로고    scopus 로고
    • Differential susceptibility to obesity between male, female and ovariectomized female mice
    • Hong J, Stubbins RE, Smith RR, Harvey AE, Nunez NP. Differential susceptibility to obesity between male, female and ovariectomized female mice. Nutr J. 2009;8:11.
    • (2009) Nutr J. , vol.8 , pp. 11
    • Hong, J.1    Stubbins, R.E.2    Smith, R.R.3    Harvey, A.E.4    Nunez, N.P.5
  • 169
    • 0036188807 scopus 로고    scopus 로고
    • Changes in proinflammatory cytokine activity after menopause
    • Pfeilschifter J, Koditz R, Pfohl M, Schatz H. Changes in proinflammatory cytokine activity after menopause. Endocr Rev. 2002;23:90-119.
    • (2002) Endocr Rev. , vol.23 , pp. 90-119
    • Pfeilschifter, J.1    Koditz, R.2    Pfohl, M.3    Schatz, H.4
  • 170
    • 0036153232 scopus 로고    scopus 로고
    • Menopause-related differences in inflammation markers and their relationship to body fat distribution and insulin-stimulated glucose disposal
    • Sites CK, Toth MJ, Cushman M, et al. Menopause-related differences in inflammation markers and their relationship to body fat distribution and insulin-stimulated glucose disposal. Fertil Steril. 2002;77:128-135.
    • (2002) Fertil Steril. , vol.77 , pp. 128-135
    • Sites, C.K.1    Toth, M.J.2    Cushman, M.3
  • 171
    • 0036084239 scopus 로고    scopus 로고
    • Effect of the ovarian hormones on GLUT4 expression and contraction-stimulated glucose uptake
    • Campbell SE, Febbraio MA. Effect of the ovarian hormones on GLUT4 expression and contraction-stimulated glucose uptake. Am J Physiol Endocrinol Metab. 2002; 282:E1139-E1146.
    • (2002) Am J Physiol Endocrinol Metab. , vol.282
    • Campbell, S.E.1    Febbraio, M.A.2
  • 172
    • 84870255429 scopus 로고    scopus 로고
    • Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance
    • Stubbins RE, Holcomb VB, Hong J, Nunez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr. 2012;51:861-870.
    • (2012) Eur J Nutr. , vol.51 , pp. 861-870
    • Stubbins, R.E.1    Holcomb, V.B.2    Hong, J.3    Nunez, N.P.4
  • 173
    • 64549157561 scopus 로고    scopus 로고
    • Estradiol stimulates Akt, AMP-activated protein kinase (AMPK) and TBC1D1/4, but not glucose uptake in rat soleus
    • Rogers NH, Witczak CA, Hirshman MF, Goodyear LJ, Greenberg AS. Estradiol stimulates Akt, AMP-activated protein kinase (AMPK) and TBC1D1/4, but not glucose uptake in rat soleus. Biochem Biophys Res Commun. 2009;382:646-650.
    • (2009) Biochem Biophys Res Commun. , vol.382 , pp. 646-650
    • Rogers, N.H.1    Witczak, C.A.2    Hirshman, M.F.3    Goodyear, L.J.4    Greenberg, A.S.5
  • 174
    • 79954461307 scopus 로고    scopus 로고
    • In vivo stimulation of oestrogen receptor α increases insulin-stimulated skeletal muscle glucose uptake
    • Gorres BK, Bomhoff GL, Morris JK, Geiger PC. In vivo stimulation of oestrogen receptor α increases insulin-stimulated skeletal muscle glucose uptake. J Physiol. 2011;589: 2041-2054.
    • (2011) J Physiol. , vol.589 , pp. 2041-2054
    • Gorres, B.K.1    Bomhoff, G.L.2    Morris, J.K.3    Geiger, P.C.4
  • 176
    • 78649331876 scopus 로고    scopus 로고
    • Acute effects of 17 β-estradiol and genistein on insulin sensitivity and spatial memory in aged ovariectomized female rats
    • Alonso A, Gonzalez-Pardo H, Garrido P, et al. Acute effects of 17 β-estradiol and genistein on insulin sensitivity and spatial memory in aged ovariectomized female rats. Age (Dordr). 2010;32:421-434.
    • (2010) Age (Dordr). , vol.32 , pp. 421-434
    • Alonso, A.1    Gonzalez-Pardo, H.2    Garrido, P.3
  • 177
    • 80052564156 scopus 로고    scopus 로고
    • Testosterone or 17β-estradiol exposure reveals sex-specific effects on glucose and lipid metabolism in human myotubes
    • Salehzadeh F, Rune A, Osler M, Al-Khalili L. Testosterone or 17β-estradiol exposure reveals sex-specific effects on glucose and lipid metabolism in human myotubes. J Endocrinol. 2011;210:219-229.
    • (2011) J Endocrinol. , vol.210 , pp. 219-229
    • Salehzadeh, F.1    Rune, A.2    Osler, M.3    Al-Khalili, L.4
  • 178
    • 36549016068 scopus 로고    scopus 로고
    • Oestradiol replacement treatment and glucose homeostasis in two men with congenital aromatase deficiency: evidence for a role of oestradiol and sex steroids imbalance on insulin sensitivity in men
    • Rochira V, Madeo B, Zirilli L, Caffagni G, Maffei L, Carani C. Oestradiol replacement treatment and glucose homeostasis in two men with congenital aromatase deficiency: evidence for a role of oestradiol and sex steroids imbalance on insulin sensitivity in men. DiabetMed. 2007; 24:1491-1495.
    • (2007) DiabetMed. , vol.24 , pp. 1491-1495
    • Rochira, V.1    Madeo, B.2    Zirilli, L.3    Caffagni, G.4    Maffei, L.5    Carani, C.6
  • 179
    • 70350046653 scopus 로고    scopus 로고
    • Metformin, estrogen replacement therapy and gonadotropin inhibition fail to improve insulin sensitivity in a girl with aromatase deficiency
    • Guercio G, Di Palma MI, Pepe C, et al. Metformin, estrogen replacement therapy and gonadotropin inhibition fail to improve insulin sensitivity in a girl with aromatase deficiency. Horm Res. 2009;72:370-376.
    • (2009) Horm Res. , vol.72 , pp. 370-376
    • Guercio, G.1    Di Palma, M.I.2    Pepe, C.3
  • 180
    • 0037291609 scopus 로고    scopus 로고
    • Progressive development of insulin resistance phenotype in male mice with complete aromatase (CYP19) deficiency
    • Takeda K, Toda K, Saibara T, et al. Progressive development of insulin resistance phenotype in male mice with complete aromatase (CYP19) deficiency. J Endocrinol. 2003;176:237-246.
    • (2003) J Endocrinol. , vol.176 , pp. 237-246
    • Takeda, K.1    Toda, K.2    Saibara, T.3
  • 181
    • 10744225853 scopus 로고    scopus 로고
    • Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment
    • Maffei L, Murata Y, Rochira V, et al. Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment. J Clin Endocrinol Metab. 2004;89:61-70.
    • (2004) J Clin Endocrinol Metab. , vol.89 , pp. 61-70
    • Maffei, L.1    Murata, Y.2    Rochira, V.3
  • 182
    • 34447515493 scopus 로고    scopus 로고
    • A novel compound heterozygous mutation of the aromatase gene in an adult man: reinforced evidence on the relationship between congenital oestrogen deficiency, adiposity and the metabolic syndrome
    • Maffei L, Rochira V, Zirilli L, et al. A novel compound heterozygous mutation of the aromatase gene in an adult man: reinforced evidence on the relationship between congenital oestrogen deficiency, adiposity and the metabolic syndrome. Clin Endocrinol (Oxf). 2007;67:218-224.
    • (2007) Clin Endocrinol (Oxf). , vol.67 , pp. 218-224
    • Maffei, L.1    Rochira, V.2    Zirilli, L.3
  • 183
    • 34548436968 scopus 로고    scopus 로고
    • Estrogen and adiposity-utilizing models of aromatase deficiency to explore the relationship
    • Jones ME, McInnes KJ, Boon WC, Simpson ER. Estrogen and adiposity-utilizing models of aromatase deficiency to explore the relationship. J Steroid Biochem Mol Biol. 2007;106:3-7.
    • (2007) J Steroid Biochem Mol Biol. , vol.106 , pp. 3-7
    • Jones, M.E.1    McInnes, K.J.2    Boon, W.C.3    Simpson, E.R.4
  • 184
    • 0028792229 scopus 로고
    • Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens
    • Morishima A, GrumbachMM,Simpson ER, Fisher C, Qin K. Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metab. 1995;80:3689-3698.
    • (1995) J Clin Endocrinol Metab. , vol.80 , pp. 3689-3698
    • Morishima, A.1    Grumbach, M.M.2    Simpson, E.R.3    Fisher, C.4    Qin, K.5
  • 185
    • 67349253060 scopus 로고    scopus 로고
    • The pancreatic β-cell as a target of estrogens and xenoestrogens: implications for blood glucose homeostasis and diabetes
    • Nadal A, Alonso-Magdalena P, Soriano S, Quesada I, Ropero AB. The pancreatic β-cell as a target of estrogens and xenoestrogens: implications for blood glucose homeostasis and diabetes. Mol Cell Endocrinol. 2009;304: 63-68.
    • (2009) Mol Cell Endocrinol. , vol.304 , pp. 63-68
    • Nadal, A.1    Alonso-Magdalena, P.2    Soriano, S.3    Quesada, I.4    Ropero, A.B.5
  • 186
    • 54849433778 scopus 로고    scopus 로고
    • Insulin resistance of pregnancy involves estrogen-induced repression of muscle GLUT4
    • Barros RP, Morani A, Moriscot A, Machado UF. Insulin resistance of pregnancy involves estrogen-induced repression of muscle GLUT4. Mol Cell Endocrinol. 2008;295: 24-31.
    • (2008) Mol Cell Endocrinol. , vol.295 , pp. 24-31
    • Barros, R.P.1    Morani, A.2    Moriscot, A.3    Machado, U.F.4
  • 187
    • 34548490716 scopus 로고    scopus 로고
    • Plasma sex steroid hormones and risk of developing type 2 diabetes in women: a prospective study
    • Ding EL, Song Y, Manson JE, Rifai N, Buring JE, Liu S. Plasma sex steroid hormones and risk of developing type 2 diabetes in women: a prospective study. Diabetologia. 2007;50:2076-2084.
    • (2007) Diabetologia. , vol.50 , pp. 2076-2084
    • Ding, E.L.1    Song, Y.2    Manson, J.E.3    Rifai, N.4    Buring, J.E.5    Liu, S.6
  • 188
    • 70449113365 scopus 로고    scopus 로고
    • The association of endogenous sex hormones, adiposity, and insulin resistance with incident diabetes in postmenopausal women
    • Kalyani RR, Franco M, Dobs AS, et al. The association of endogenous sex hormones, adiposity, and insulin resistance with incident diabetes in postmenopausal women. J Clin Endocrinol Metab. 2009;94:4127-4135.
    • (2009) J Clin Endocrinol Metab. , vol.94 , pp. 4127-4135
    • Kalyani, R.R.1    Franco, M.2    Dobs, A.S.3
  • 189
    • 20144370690 scopus 로고    scopus 로고
    • Expression of oestrogen receptor α and β is higher in skeletal muscle of highly endurance-trained than of moderately active men
    • Wiik A, Gustafsson T, Esbjornsson M, et al. Expression of oestrogen receptor α and β is higher in skeletal muscle of highly endurance-trained than of moderately active men. Acta Physiol Scand. 2005;184:105-112.
    • (2005) Acta Physiol Scand. , vol.184 , pp. 105-112
    • Wiik, A.1    Gustafsson, T.2    Esbjornsson, M.3
  • 190
    • 77956320263 scopus 로고    scopus 로고
    • Estrogen regulates estrogen receptors and antioxidant gene expression in mouse skeletal muscle
    • Baltgalvis KA, Greising SM, Warren GL, Lowe DA. Estrogen regulates estrogen receptors and antioxidant gene expression in mouse skeletal muscle. PLoS One. 2010;5: e10164.
    • (2010) PLoS One. , vol.5
    • Baltgalvis, K.A.1    Greising, S.M.2    Warren, G.L.3    Lowe, D.A.4
  • 191
    • 77949911881 scopus 로고    scopus 로고
    • Genetic evidence that raised sex hormone binding globulin (SHBG) levels reduce the risk of type 2 diabetes
    • Perry JR, Weedon MN, Langenberg C, et al. Genetic evidence that raised sex hormone binding globulin (SHBG) levels reduce the risk of type 2 diabetes. Hum Mol Genet. 2010;19:535-544.
    • (2010) Hum Mol Genet. , vol.19 , pp. 535-544
    • Perry, J.R.1    Weedon, M.N.2    Langenberg, C.3
  • 192
    • 74949093473 scopus 로고    scopus 로고
    • Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERα-deficient mice
    • Ribas V, Nguyen MT, Henstridge DC, et al. Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERα-deficient mice. Am J Physiol Endocrinol Metab. 2010;298:E304-E319.
    • (2010) Am J Physiol Endocrinol Metab. , vol.298
    • Ribas, V.1    Nguyen, M.T.2    Henstridge, D.C.3
  • 193
    • 66449105068 scopus 로고    scopus 로고
    • Estrogens protect against high-fat diet-induced insulin resistance and glucose intolerance in mice
    • Riant E, Waget A, Cogo H, Arnal JF, Burcelin R, Gourdy P. Estrogens protect against high-fat diet-induced insulin resistance and glucose intolerance in mice. Endocrinology. 2009;150:2109-2117.
    • (2009) Endocrinology. , vol.150 , pp. 2109-2117
    • Riant, E.1    Waget, A.2    Cogo, H.3    Arnal, J.F.4    Burcelin, R.5    Gourdy, P.6
  • 194
    • 33244495940 scopus 로고    scopus 로고
    • Evidence that oestrogen receptor-α plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver
    • Bryzgalova G, Gao H, Ahren B, et al. Evidence that oestrogen receptor-α plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver. Diabetologia. 2006;49:588-597.
    • (2006) Diabetologia. , vol.49 , pp. 588-597
    • Bryzgalova, G.1    Gao, H.2    Ahren, B.3
  • 195
    • 84892625032 scopus 로고    scopus 로고
    • Skeletal muscle specific ERα deletion is causal for the metabolic syndrome
    • Ribas V, Drew BG, Soleymani T, Daraei P, Hevener A. Skeletal muscle specific ERα deletion is causal for the metabolic syndrome. Endocr Rev. 2010;31:S5.
    • (2010) Endocr Rev. , vol.31
    • Ribas, V.1    Drew, B.G.2    Soleymani, T.3    Daraei, P.4    Hevener, A.5
  • 197
    • 0041852780 scopus 로고    scopus 로고
    • 17β-Estradiol upregulates the expression of peroxisome proliferator-activated receptor α and lipid oxidative genes in skeletal muscle
    • Campbell SE, Mehan KA, Tunstall RJ, Febbraio MA, Cameron-Smith D. 17β-Estradiol upregulates the expression of peroxisome proliferator-activated receptor α and lipid oxidative genes in skeletal muscle. J Mol Endocrinol. 2003;31:37-45.
    • (2003) J Mol Endocrinol. , vol.31 , pp. 37-45
    • Campbell, S.E.1    Mehan, K.A.2    Tunstall, R.J.3    Febbraio, M.A.4    Cameron-Smith, D.5
  • 198
    • 67650663164 scopus 로고    scopus 로고
    • 17β-Estradiol treatment is unable to reproduce p85 α redistribution associated with gestational insulin resistance in rats
    • Alonso A, Ordonez P, Fernandez R, et al. 17β-Estradiol treatment is unable to reproduce p85 α redistribution associated with gestational insulin resistance in rats. J Steroid Biochem Mol Biol. 2009;116:160-170.
    • (2009) J Steroid Biochem Mol Biol. , vol.116 , pp. 160-170
    • Alonso, A.1    Ordonez, P.2    Fernandez, R.3
  • 199
    • 0029920870 scopus 로고    scopus 로고
    • Effects of ovariectomy and exercise training on muscle GLUT-4 content and glucose metabolism in rats
    • Hansen PA, McCarthy TJ, Pasia EN, Spina RJ, Gulve EA. Effects of ovariectomy and exercise training on muscle GLUT-4 content and glucose metabolism in rats. J Appl Physiol. 1996;80:1605-1611.
    • (1996) J Appl Physiol. , vol.80 , pp. 1605-1611
    • Hansen, P.A.1    McCarthy, T.J.2    Pasia, E.N.3    Spina, R.J.4    Gulve, E.A.5
  • 200
    • 80052742071 scopus 로고    scopus 로고
    • Estrogen receptors and the metabolic network
    • Barros RP, Gustafsson JA. Estrogen receptors and the metabolic network. Cell Metab. 2011;14:289-299.
    • (2011) Cell Metab. , vol.14 , pp. 289-299
    • Barros, R.P.1    Gustafsson, J.A.2
  • 201
    • 69949102948 scopus 로고    scopus 로고
    • Multiple signalling pathways redundantly control glucose transporter GLUT4 gene transcription in skeletal muscle
    • Murgia M, Jensen TE, Cusinato M, Garcia M, Richter EA, Schiaffino S. Multiple signalling pathways redundantly control glucose transporter GLUT4 gene transcription in skeletal muscle. J Physiol. 2009;587:4319-4327.
    • (2009) J Physiol. , vol.587 , pp. 4319-4327
    • Murgia, M.1    Jensen, T.E.2    Cusinato, M.3    Garcia, M.4    Richter, E.A.5    Schiaffino, S.6
  • 202
    • 13244281673 scopus 로고    scopus 로고
    • Mechanisms regulating GLUT4 glucose transporter expression and glucose transport in skeletal muscle
    • Zorzano A, Palacin M, Guma A. Mechanisms regulating GLUT4 glucose transporter expression and glucose transport in skeletal muscle. Acta Physiol Scand. 2005;183: 43-58.
    • (2005) Acta Physiol Scand. , vol.183 , pp. 43-58
    • Zorzano, A.1    Palacin, M.2    Guma, A.3
  • 203
    • 73649091452 scopus 로고    scopus 로고
    • Exercise, sex, menstrual cycle phase, and 17β-estradiol influence metabolism-related genes in human skeletal muscle
    • Fu MH, Maher AC, Hamadeh MJ, Ye C, Tarnopolsky MA. Exercise, sex, menstrual cycle phase, and 17β-estradiol influence metabolism-related genes in human skeletal muscle. Physiol Genomics. 2009;40:34-47.
    • (2009) Physiol Genomics. , vol.40 , pp. 34-47
    • Fu, M.H.1    Maher, A.C.2    Hamadeh, M.J.3    Ye, C.4    Tarnopolsky, M.A.5
  • 204
    • 69749085980 scopus 로고    scopus 로고
    • Higher intramuscular triacylglycerol in women does not impair insulin sensitivity and proximal insulin signaling
    • Hoeg L, Roepstorff C, Thiele M, Richter EA, Wojtaszewski JF, Kiens B. Higher intramuscular triacylglycerol in women does not impair insulin sensitivity and proximal insulin signaling. J Appl Physiol. 2009;107:824-831.
    • (2009) J Appl Physiol. , vol.107 , pp. 824-831
    • Hoeg, L.1    Roepstorff, C.2    Thiele, M.3    Richter, E.A.4    Wojtaszewski, J.F.5    Kiens, B.6
  • 205
    • 0032104120 scopus 로고    scopus 로고
    • Evidence for defects in the trafficking and translocation of GLUT4 glucose transporters in skeletal muscle as a cause of human insulin resistance
    • Garvey WT, Maianu L, Zhu JH, Brechtel-Hook G, Wallace P, Baron AD. Evidence for defects in the trafficking and translocation of GLUT4 glucose transporters in skeletal muscle as a cause of human insulin resistance. J Clin Invest. 1998;101:2377-2386.
    • (1998) J Clin Invest. , vol.101 , pp. 2377-2386
    • Garvey, W.T.1    Maianu, L.2    Zhu, J.H.3    Brechtel-Hook, G.4    Wallace, P.5    Baron, A.D.6
  • 206
    • 0026723385 scopus 로고
    • Gene expression of GLUT4 in skeletal muscle from insulin-resistant patients with obesity, IGT, GDM, and NIDDM
    • Garvey WT, Maianu L, Hancock JA, Golichowski AM, Baron A. Gene expression of GLUT4 in skeletal muscle from insulin-resistant patients with obesity, IGT, GDM, and NIDDM. Diabetes. 1992;41:465-475.
    • (1992) Diabetes. , vol.41 , pp. 465-475
    • Garvey, W.T.1    Maianu, L.2    Hancock, J.A.3    Golichowski, A.M.4    Baron, A.5
  • 207
    • 0026953735 scopus 로고
    • Muscle glucose transport, GLUT-4 content, and degree of exercise training in obese Zucker rats
    • Banks EA, Brozinick JT Jr, Yaspelkis BB III, Kang HY, Ivy JL. Muscle glucose transport, GLUT-4 content, and degree of exercise training in obese Zucker rats. Am J Physiol. 1992;263:E1010-E1015.
    • (1992) Am J Physiol. , vol.263
    • Banks, E.A.1    Brozinick Jr., J.T.2    Yaspelkis, B.B.3    Kang, H.Y.4    Ivy, J.L.5
  • 208
    • 0027425174 scopus 로고
    • Effects of exercise training on muscle GLUT-4 protein content and translocation in obese Zucker rats
    • Brozinick JT Jr, Etgen GJ Jr, Yaspelkis BB III, Kang HY, Ivy JL. Effects of exercise training on muscle GLUT-4 protein content and translocation in obese Zucker rats. Am J Physiol. 1993;265:E419-E427.
    • (1993) Am J Physiol. , vol.265
    • Brozinick Jr., J.T.1    Etgen Jr., G.J.2    Yaspelkis, B.B.3    Kang, H.Y.4    Ivy, J.L.5
  • 209
    • 0028026320 scopus 로고
    • Glucose uptake and GLUT-4 protein distribution in skeletal muscle of the obese Zucker rat
    • Brozinick JT Jr, Etgen GJ Jr, Yaspelkis BB III, Ivy JL. Glucose uptake and GLUT-4 protein distribution in skeletal muscle of the obese Zucker rat. Am J Physiol. 1994;267: R236-R243.
    • (1994) Am J Physiol. , vol.267
    • Brozinick Jr., J.T.1    Etgen Jr., G.J.2    Yaspelkis, B.B.3    Ivy, J.L.4
  • 210
    • 0033645010 scopus 로고    scopus 로고
    • Exercise and thiazolidinedione therapy normalize insulin action in the obese Zucker fatty rat
    • Hevener AL, Reichart D, Olefsky J. Exercise and thiazolidinedione therapy normalize insulin action in the obese Zucker fatty rat. Diabetes. 2000;49:2154-2159.
    • (2000) Diabetes. , vol.49 , pp. 2154-2159
    • Hevener, A.L.1    Reichart, D.2    Olefsky, J.3
  • 211
    • 0028200755 scopus 로고
    • Physical training increases muscleGLUT4protein andmRNAin patients with NIDDM
    • Dela F, Ploug T, Handberg A, et al. Physical training increases muscleGLUT4protein andmRNAin patients with NIDDM. Diabetes. 1994;43:862-865.
    • (1994) Diabetes. , vol.43 , pp. 862-865
    • Dela, F.1    Ploug, T.2    Handberg, A.3
  • 212
    • 0025094347 scopus 로고
    • Effects of exercise training on insulin-regulatable glucose-transporter protein levels in rat skeletal muscle
    • Rodnick KJ, Holloszy JO, Mondon CE, James DE. Effects of exercise training on insulin-regulatable glucose-transporter protein levels in rat skeletal muscle. Diabetes. 1990; 39:1425-1429.
    • (1990) Diabetes. , vol.39 , pp. 1425-1429
    • Rodnick, K.J.1    Holloszy, J.O.2    Mondon, C.E.3    James, D.E.4
  • 213
    • 0035991422 scopus 로고    scopus 로고
    • Effect of endurance training on oestrogen receptorα expression in different rat skeletal muscle type
    • Lemoine S, Granier P, Tiffoche C, et al. Effect of endurance training on oestrogen receptorα expression in different rat skeletal muscle type. Acta Physiol Scand. 2002;175:211-217.
    • (2002) Acta Physiol Scand. , vol.175 , pp. 211-217
    • Lemoine, S.1    Granier, P.2    Tiffoche, C.3
  • 214
    • 0036203152 scopus 로고    scopus 로고
    • Effect of endurance training on oestrogen receptor α transcripts in rat skeletal muscle
    • Lemoine S, Granier P, Tiffoche C, et al. Effect of endurance training on oestrogen receptor α transcripts in rat skeletal muscle. Acta Physiol Scand. 2002;174:283-289.
    • (2002) Acta Physiol Scand. , vol.174 , pp. 283-289
    • Lemoine, S.1    Granier, P.2    Tiffoche, C.3
  • 215
    • 0034717280 scopus 로고    scopus 로고
    • The MEF2A isoform is required for striated muscle-specific expression of the insulin-responsive GLUT4 glucose transporter
    • Mora S, Pessin JE. The MEF2A isoform is required for striated muscle-specific expression of the insulin-responsive GLUT4 glucose transporter. J Biol Chem. 2000;275: 16323-16328.
    • (2000) J Biol Chem. , vol.275 , pp. 16323-16328
    • Mora, S.1    Pessin, J.E.2
  • 216
    • 74049129003 scopus 로고    scopus 로고
    • Myocyte enhancer factor 2 and class II histone deacetylases control a gender-specific pathway of cardioprotection mediated by the estrogen receptor
    • van Rooij E, Fielitz J, Sutherland LB, et al. Myocyte enhancer factor 2 and class II histone deacetylases control a gender-specific pathway of cardioprotection mediated by the estrogen receptor. Circ Res. 2010;106:155-165.
    • (2010) Circ Res. , vol.106 , pp. 155-165
    • van Rooij, E.1    Fielitz, J.2    Sutherland, L.B.3
  • 217
    • 0142039986 scopus 로고    scopus 로고
    • Differential regulation of the muscle-specific GLUT4 enhancer in regenerating and adult skeletal muscle
    • Moreno H, Serrano AL, Santalucia T, et al. Differential regulation of the muscle-specific GLUT4 enhancer in regenerating and adult skeletal muscle. J Biol Chem. 2003; 278:40557-40564.
    • (2003) J Biol Chem. , vol.278 , pp. 40557-40564
    • Moreno, H.1    Serrano, A.L.2    Santalucia, T.3
  • 218
    • 84255186061 scopus 로고    scopus 로고
    • The nuclear receptor PPARβ/δ programs muscle glucose metabolism in cooperation with AMPK and MEF2
    • Gan Z, Burkart-Hartman EM, Han DH, et al. The nuclear receptor PPARβ/δ programs muscle glucose metabolism in cooperation with AMPK and MEF2. Genes Dev. 2011; 25:2619-2630.
    • (2011) Genes Dev. , vol.25 , pp. 2619-2630
    • Gan, Z.1    Burkart-Hartman, E.M.2    Han, D.H.3
  • 219
    • 0034604547 scopus 로고    scopus 로고
    • Identification of a 30-base pair regulatory element and novel DNA binding protein that regulates the human GLUT4 promoter in transgenic mice
    • Oshel KM, Knight JB, Cao KT, Thai MV, Olson AL. Identification of a 30-base pair regulatory element and novel DNA binding protein that regulates the human GLUT4 promoter in transgenic mice. J Biol Chem. 2000;275: 23666-23673.
    • (2000) J Biol Chem. , vol.275 , pp. 23666-23673
    • Oshel, K.M.1    Knight, J.B.2    Cao, K.T.3    Thai, M.V.4    Olson, A.L.5
  • 220
    • 53149104368 scopus 로고    scopus 로고
    • CaMK activation during exercise is required for histone hyperacetylation and MEF2A binding at the MEF2 site on the Glut4 gene
    • Smith JA, Kohn TA, Chetty AK, Ojuka EO. CaMK activation during exercise is required for histone hyperacetylation and MEF2A binding at the MEF2 site on the Glut4 gene. Am J Physiol Endocrinol Metab. 2008;295:E698-E704.
    • (2008) Am J Physiol Endocrinol Metab. , vol.295
    • Smith, J.A.1    Kohn, T.A.2    Chetty, A.K.3    Ojuka, E.O.4
  • 221
    • 79961021758 scopus 로고    scopus 로고
    • EF2Abinding to the Glut4 promoter occurs via an AMPKα2-dependent mechanism
    • Gong H, Xie J, Zhang N, Yao L, Zhang Y.MEF2Abinding to the Glut4 promoter occurs via an AMPKα2-dependent mechanism. Med Sci Sports Exerc. 2011;43:1441-1450.
    • (2011) Med Sci Sports Exerc. , vol.43 , pp. 1441-1450
    • Gong, H.1    Xie, J.2    Zhang, N.3    Yao, L.4    Zhang, Y.M.5
  • 224
    • 2042425906 scopus 로고    scopus 로고
    • The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibitingFOXOtranscription factors
    • Stitt TN, Drujan D, Clarke BA, et al. The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibitingFOXOtranscription factors. Mol Cell. 2004;14:395-403.
    • (2004) Mol Cell. , vol.14 , pp. 395-403
    • Stitt, T.N.1    Drujan, D.2    Clarke, B.A.3
  • 225
    • 48749102153 scopus 로고    scopus 로고
    • Oestrogen receptors mediate oestrogen-induced increases in post-exercise rat skeletal muscle satellite cells
    • Enns DL, Iqbal S, Tiidus PM. Oestrogen receptors mediate oestrogen-induced increases in post-exercise rat skeletal muscle satellite cells. Acta Physiol (Oxf). 2008;194:81-93.
    • (2008) Acta Physiol (Oxf). , vol.194 , pp. 81-93
    • Enns, D.L.1    Iqbal, S.2    Tiidus, P.M.3
  • 226
    • 39049131254 scopus 로고    scopus 로고
    • Estrogen influences satellite cell activation and proliferation following downhill running in rats
    • Enns DL, Tiidus PM. Estrogen influences satellite cell activation and proliferation following downhill running in rats. J Appl Physiol. 2008;104:347-353.
    • (2008) J Appl Physiol. , vol.104 , pp. 347-353
    • Enns, D.L.1    Tiidus, P.M.2
  • 227
    • 71649103267 scopus 로고    scopus 로고
    • Oestrogen receptor-α activation augments post-exercise myoblast proliferation
    • Thomas A, Bunyan K, Tiidus PM. Oestrogen receptor-α activation augments post-exercise myoblast proliferation. Acta Physiol (Oxf). 2010;198:81-89.
    • (2010) Acta Physiol (Oxf). , vol.198 , pp. 81-89
    • Thomas, A.1    Bunyan, K.2    Tiidus, P.M.3
  • 228
    • 77954861858 scopus 로고    scopus 로고
    • Effect of estradiol-17β on protein synthesis and degradation rates in fused bovine satellite cell cultures
    • Kamanga-Sollo E, White ME, Hathaway MR, Weber WJ, Dayton WR. Effect of estradiol-17β on protein synthesis and degradation rates in fused bovine satellite cell cultures. Domest Anim Endocrinol. 2010;39:54-62.
    • (2010) Domest Anim Endocrinol. , vol.39 , pp. 54-62
    • Kamanga-Sollo, E.1    White, M.E.2    Hathaway, M.R.3    Weber, W.J.4    Dayton, W.R.5
  • 229
    • 25644447454 scopus 로고    scopus 로고
    • Upregulation of PI3K/Akt signaling by 17β-estradiol through activation of estrogen receptor-α, but not estrogen receptor-β, and stimulates cell growth in breast cancer cells
    • Lee YR, Park J, Yu HN, Kim JS, Youn HJ, Jung SH. Upregulation of PI3K/Akt signaling by 17β-estradiol through activation of estrogen receptor-α, but not estrogen receptor-β, and stimulates cell growth in breast cancer cells. Biochem Biophys Res Commun. 2005;336:1221-1226.
    • (2005) Biochem Biophys Res Commun. , vol.336 , pp. 1221-1226
    • Lee, Y.R.1    Park, J.2    Yu, H.N.3    Kim, J.S.4    Youn, H.J.5    Jung, S.H.6
  • 230
    • 79551699799 scopus 로고    scopus 로고
    • Estrogen receptor α induces down-regulation of PTEN through PI3-kinase activation in breast cancer cells
    • Noh EM, Lee YR, Chay KO, et al. Estrogen receptor α induces down-regulation of PTEN through PI3-kinase activation in breast cancer cells. Mol Med Report. 2011;4: 215-219.
    • (2011) Mol Med Report. , vol.4 , pp. 215-219
    • Noh, E.M.1    Lee, Y.R.2    Chay, K.O.3
  • 231
    • 0034727094 scopus 로고    scopus 로고
    • Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase
    • Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK. Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature. 2000;407:538-541.
    • (2000) Nature. , vol.407 , pp. 538-541
    • Simoncini, T.1    Hafezi-Moghadam, A.2    Brazil, D.P.3    Ley, K.4    Chin, W.W.5    Liao, J.K.6
  • 233
    • 33748681724 scopus 로고    scopus 로고
    • Estrogen receptor protein interaction with phosphatidylinositol 3-kinase leads to activation of phosphorylated Akt and extracellular signal-regulated kinase 1/2 in the same population of cortical neurons: a unified mechanism of estrogen action
    • Mannella P, Brinton RD. Estrogen receptor protein interaction with phosphatidylinositol 3-kinase leads to activation of phosphorylated Akt and extracellular signal-regulated kinase 1/2 in the same population of cortical neurons: a unified mechanism of estrogen action. J Neurosci. 2006; 26:9439-9447.
    • (2006) J Neurosci. , vol.26 , pp. 9439-9447
    • Mannella, P.1    Brinton, R.D.2
  • 234
    • 77955076056 scopus 로고    scopus 로고
    • Extracellularregulated kinase and p38 mitogen-activated protein kinases are involved in the antiapoptotic action of 17β-estradiol in skeletal muscle cells
    • Ronda AC, Vasconsuelo A, Boland R. Extracellularregulated kinase and p38 mitogen-activated protein kinases are involved in the antiapoptotic action of 17β-estradiol in skeletal muscle cells. J Endocrinol. 2010;206: 235-246.
    • (2010) J Endocrinol. , vol.206 , pp. 235-246
    • Ronda, A.C.1    Vasconsuelo, A.2    Boland, R.3
  • 235
    • 77957289109 scopus 로고    scopus 로고
    • Role of estrogen receptors, PKC and Src in ERK2 and p38 MAPK signaling triggered by 17β-estradiol in skeletal muscle cells
    • Ronda AC, Buitrago C, Boland R. Role of estrogen receptors, PKC and Src in ERK2 and p38 MAPK signaling triggered by 17β-estradiol in skeletal muscle cells. J Steroid Biochem Mol Biol. 2010;122:287-294.
    • (2010) J Steroid Biochem Mol Biol. , vol.122 , pp. 287-294
    • Ronda, A.C.1    Buitrago, C.2    Boland, R.3
  • 236
    • 0038381442 scopus 로고    scopus 로고
    • Maturation of the regulation of GLUT4 activity by p38 MAPK during L6 cell myogenesis
    • Niu W, Huang C, Nawaz Z, et al. Maturation of the regulation of GLUT4 activity by p38 MAPK during L6 cell myogenesis. J Biol Chem. 2003;278:17953-17962.
    • (2003) J Biol Chem. , vol.278 , pp. 17953-17962
    • Niu, W.1    Huang, C.2    Nawaz, Z.3
  • 238
    • 0033537853 scopus 로고    scopus 로고
    • An inhibitor of p38 mitogen-activated protein kinase prevents insulin-stimulated glucose transport but not glucose transporter translocation in 3T3-L1 adipocytes and L6 myotubes
    • Sweeney G, Somwar R, Ramlal T, Volchuk A, Ueyama A, Klip A. An inhibitor of p38 mitogen-activated protein kinase prevents insulin-stimulated glucose transport but not glucose transporter translocation in 3T3-L1 adipocytes and L6 myotubes. J Biol Chem. 1999;274:10071-10078.
    • (1999) J Biol Chem. , vol.274 , pp. 10071-10078
    • Sweeney, G.1    Somwar, R.2    Ramlal, T.3    Volchuk, A.4    Ueyama, A.5    Klip, A.6
  • 239
    • 0039567273 scopus 로고    scopus 로고
    • Obesity and sarcopenia after menopause are reversed by sex hormone replacement therapy
    • Sorensen MB, Rosenfalck AM, Hojgaard L, Ottesen B. Obesity and sarcopenia after menopause are reversed by sex hormone replacement therapy. Obes Res. 2001;9:622-626.
    • (2001) Obes Res. , vol.9 , pp. 622-626
    • Sorensen, M.B.1    Rosenfalck, A.M.2    Hojgaard, L.3    Ottesen, B.4
  • 240
    • 39449100832 scopus 로고    scopus 로고
    • Humansarcopenia reveals an increase inSOCS-3and myostatin and a reduced efficiency of Akt phosphorylation
    • Leger B, Derave W, De Bock K, Hespel P, Russell AP. Humansarcopenia reveals an increase inSOCS-3and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Res. 2008;11:163-175B.
    • (2008) Rejuvenation Res. , vol.11
    • Leger, B.1    Derave, W.2    De Bock, K.3    Hespel, P.4    Russell, A.P.5
  • 241
    • 0034251875 scopus 로고    scopus 로고
    • Estrogen and gender effects on muscle damage, inflammation, and oxidative stress
    • Tiidus PM. Estrogen and gender effects on muscle damage, inflammation, and oxidative stress. Can J Appl Physiol. 2000;25:274-287.
    • (2000) Can J Appl Physiol. , vol.25 , pp. 274-287
    • Tiidus, P.M.1
  • 243
    • 27244443774 scopus 로고    scopus 로고
    • Postmenopausal hormone therapy and body composition-a substudy of the estrogen plus progestin trial of the Women's Health Initiative
    • Chen Z, Bassford T, Green SB, et al. Postmenopausal hormone therapy and body composition-a substudy of the estrogen plus progestin trial of the Women's Health Initiative. Am J Clin Nutr. 2005;82:651-656.
    • (2005) Am J Clin Nutr. , vol.82 , pp. 651-656
    • Chen, Z.1    Bassford, T.2    Green, S.B.3
  • 244
    • 0034862882 scopus 로고    scopus 로고
    • Effects of hormone replacement therapy and high-impact physical exercise on skeletal muscle in postmenopausal women: a randomized placebo-controlled study
    • Sipila S, Taaffe DR, Cheng S, Puolakka J, Toivanen J, Suominen H. Effects of hormone replacement therapy and high-impact physical exercise on skeletal muscle in postmenopausal women: a randomized placebo-controlled study. Clin Sci (Lond). 2001;101:147-157.
    • (2001) Clin Sci (Lond). , vol.101 , pp. 147-157
    • Sipila, S.1    Taaffe, D.R.2    Cheng, S.3    Puolakka, J.4    Toivanen, J.5    Suominen, H.6
  • 245
    • 0037387162 scopus 로고    scopus 로고
    • Resistance training in postmenopausal women with and without hormone therapy
    • Teixeira PJ, Going SB, Houtkooper LB, et al. Resistance training in postmenopausal women with and without hormone therapy. Med Sci Sports Exerc. 2003;35:555-562.
    • (2003) Med Sci Sports Exerc. , vol.35 , pp. 555-562
    • Teixeira, P.J.1    Going, S.B.2    Houtkooper, L.B.3
  • 247
    • 33750296520 scopus 로고    scopus 로고
    • Soleus muscle force following downhill running in ovariectomized rats treated with estrogen
    • Sotiriadou S, Kyparos A, Albani M, et al. Soleus muscle force following downhill running in ovariectomized rats treated with estrogen. Appl Physiol Nutr Metab. 2006;31: 449-459.
    • (2006) Appl Physiol Nutr Metab. , vol.31 , pp. 449-459
    • Sotiriadou, S.1    Kyparos, A.2    Albani, M.3
  • 248
    • 44549086906 scopus 로고    scopus 로고
    • 17β-Estradiol signaling in skeletal muscle cells and its relationship to apoptosis
    • Boland R, Vasconsuelo A, Milanesi L, Ronda AC, de Boland AR. 17β-Estradiol signaling in skeletal muscle cells and its relationship to apoptosis. Steroids. 2008;73:859-863.
    • (2008) Steroids. , vol.73 , pp. 859-863
    • Boland, R.1    Vasconsuelo, A.2    Milanesi, L.3    Ronda, A.C.4    de Boland, A.R.5
  • 249
    • 39749089950 scopus 로고    scopus 로고
    • 17β-Estradiol abrogates apoptosis in murine skeletal muscle cells through estrogen receptors: role of the phosphatidylinositol 3-kinase/Akt pathway
    • Vasconsuelo A, Milanesi L, Boland R. 17β-Estradiol abrogates apoptosis in murine skeletal muscle cells through estrogen receptors: role of the phosphatidylinositol 3-kinase/Akt pathway. J Endocrinol. 2008;196:385-397.
    • (2008) J Endocrinol. , vol.196 , pp. 385-397
    • Vasconsuelo, A.1    Milanesi, L.2    Boland, R.3
  • 250
    • 77951499165 scopus 로고    scopus 로고
    • Female adult mouse cardiomyocytes are protected against oxidative stress
    • Wang F, He Q, Sun Y, Dai X, Yang XP. Female adult mouse cardiomyocytes are protected against oxidative stress. Hypertension. 2010;55:1172-1178.
    • (2010) Hypertension. , vol.55 , pp. 1172-1178
    • Wang, F.1    He, Q.2    Sun, Y.3    Dai, X.4    Yang, X.P.5
  • 252
    • 0036875643 scopus 로고    scopus 로고
    • Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor α (ERα): a potential role for estrogen receptor β (ERβ)
    • Naaz A, Zakroczymski M, Heine P, et al. Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor α (ERα): a potential role for estrogen receptor β (ERβ). Horm Metab Res. 2002;34:758-763.
    • (2002) Horm Metab Res. , vol.34 , pp. 758-763
    • Naaz, A.1    Zakroczymski, M.2    Heine, P.3
  • 254
    • 78751477559 scopus 로고    scopus 로고
    • Lipid-induced insulin resistance affects women less than men and is not accompanied by inflammation or impaired proximal insulin signaling
    • Hoeg LD, Sjoberg KA, Jeppesen J, et al. Lipid-induced insulin resistance affects women less than men and is not accompanied by inflammation or impaired proximal insulin signaling. Diabetes. 2011;60:64-73.
    • (2011) Diabetes. , vol.60 , pp. 64-73
    • Hoeg, L.D.1    Sjoberg, K.A.2    Jeppesen, J.3
  • 255
    • 0034251864 scopus 로고    scopus 로고
    • Sex differences in substrate metabolism and energy homeostasis
    • Cortright RN, Koves TR. Sex differences in substrate metabolism and energy homeostasis. Can J Appl Physiol. 2000;25:288-311.
    • (2000) Can J Appl Physiol. , vol.25 , pp. 288-311
    • Cortright, R.N.1    Koves, T.R.2
  • 256
    • 80053403320 scopus 로고    scopus 로고
    • Skeletal muscle triglycerides, diacylglycerols, and ceramides in insulin resistance: another paradox in endurance-trained athletes?
    • Amati F, Dube JJ, Alvarez-Carnero E, et al. Skeletal muscle triglycerides, diacylglycerols, and ceramides in insulin resistance: another paradox in endurance-trained athletes? Diabetes. 2011;60:2588-2597.
    • (2011) Diabetes , vol.60 , pp. 2588-2597
    • Amati, F.1    Dube, J.J.2    Alvarez-Carnero, E.3
  • 257
    • 77957778868 scopus 로고    scopus 로고
    • Women have higher protein content of β-oxidation enzymes in skeletal muscle than men
    • Maher AC, Akhtar M, Vockley J, Tarnopolsky MA. Women have higher protein content of β-oxidation enzymes in skeletal muscle than men. PLoS One. 2010;5: e12025.
    • (2010) PLoS One. , vol.5
    • Maher, A.C.1    Akhtar, M.2    Vockley, J.3    Tarnopolsky, M.A.4
  • 258
    • 0026773244 scopus 로고
    • Increased plasma FFA uptake and oxidation during prolonged exercise in trained vs
    • Turcotte LP, Richter EA, Kiens B. Increased plasma FFA uptake and oxidation during prolonged exercise in trained vs. untrained humans. Am J Physiol. 1992;262:E791-E799.
    • (1992) untrained humans. Am J Physiol. , vol.262
    • Turcotte, L.P.1    Richter, E.A.2    Kiens, B.3
  • 259
    • 21244444322 scopus 로고    scopus 로고
    • Estrogen supplementation reduces whole body leucine and carbohydrate oxidation and increases lipid oxidation in men during endurance exercise
    • Hamadeh MJ, Devries MC, Tarnopolsky MA. Estrogen supplementation reduces whole body leucine and carbohydrate oxidation and increases lipid oxidation in men during endurance exercise. J Clin Endocrinol Metab. 2005; 90:3592-3599.
    • (2005) J Clin Endocrinol Metab. , vol.90 , pp. 3592-3599
    • Hamadeh, M.J.1    Devries, M.C.2    Tarnopolsky, M.A.3
  • 260
    • 77955748551 scopus 로고    scopus 로고
    • Men supplemented with 17β-estradiol have increased β-oxidation capacity in skeletal muscle
    • Maher AC, Akhtar M, Tarnopolsky MA. Men supplemented with 17β-estradiol have increased β-oxidation capacity in skeletal muscle. Physiol Genomics. 2010;42:342-347.
    • (2010) Physiol Genomics. , vol.42 , pp. 342-347
    • Maher, A.C.1    Akhtar, M.2    Tarnopolsky, M.A.3
  • 261
    • 44449161505 scopus 로고    scopus 로고
    • Estradiol and the estradiol metabolite, 2-hydroxyestradiol, activate AMP-activated protein kinase in C2C12 myotubes
    • D'Eon TM, Rogers NH, Stancheva ZS, Greenberg AS. Estradiol and the estradiol metabolite, 2-hydroxyestradiol, activate AMP-activated protein kinase in C2C12 myotubes. Obesity (Silver Spring). 2008;16:1284-1288.
    • (2008) Obesity (Silver Spring). , vol.16 , pp. 1284-1288
    • D'Eon, T.M.1    Rogers, N.H.2    Stancheva, Z.S.3    Greenberg, A.S.4
  • 262
    • 80052511813 scopus 로고    scopus 로고
    • The AMPK signalling pathway coordinates cell growth, autophagy and metabolism
    • Mihaylova MM, Shaw RJ. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol. 2011;13:1016-1023.
    • (2011) Nat Cell Biol. , vol.13 , pp. 1016-1023
    • Mihaylova, M.M.1    Shaw, R.J.2
  • 263
    • 80053035284 scopus 로고    scopus 로고
    • AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function
    • Hardie DG. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011;25:1895-1908.
    • (2011) Genes Dev. , vol.25 , pp. 1895-1908
    • Hardie, D.G.1
  • 264
    • 77956400845 scopus 로고    scopus 로고
    • Parenteral 17β-estradiol decreases fasting blood glucose levels in non-obese mice with short-term ovariectomy
    • Kim JY, Jo KJ, Kim OS, et al. Parenteral 17β-estradiol decreases fasting blood glucose levels in non-obese mice with short-term ovariectomy. Life Sci. 2010;87:358-366. 265. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111-1119.
    • (2010) Life Sci. , vol.87 , pp. 358-366
    • Kim, J.Y.1    Jo, K.J.2    Kim, O.S.3
  • 265
    • 18244370762 scopus 로고    scopus 로고
    • Inflammation, stress, and diabetes
    • Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111-1119.
    • (2005) J Clin Invest. , vol.115 , pp. 1111-1119
    • Wellen, K.E.1    Hotamisligil, G.S.2
  • 266
    • 0036300538 scopus 로고    scopus 로고
    • Lipidinduced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IκB-α
    • Itani SI, Ruderman NB, Schmieder F, Boden G. Lipidinduced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IκB-α. Diabetes. 2002;51:2005-2011.
    • (2002) Diabetes. , vol.51 , pp. 2005-2011
    • Itani, S.I.1    Ruderman, N.B.2    Schmieder, F.3    Boden, G.4
  • 267
    • 0346729968 scopus 로고    scopus 로고
    • Ceramide content is increased in skeletal muscle from obese insulinresistant humans
    • Adams JM II, Pratipanawatr T, Berria R, et al. Ceramide content is increased in skeletal muscle from obese insulinresistant humans. Diabetes. 2004;53:25-31.
    • (2004) Diabetes. , vol.53 , pp. 25-31
    • Adams, J.M.1    Pratipanawatr, T.2    Berria, R.3
  • 269
    • 31644443192 scopus 로고    scopus 로고
    • Ceramides in insulin resistance and lipotoxicity
    • Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res. 2006;45:42-72.
    • (2006) Prog Lipid Res. , vol.45 , pp. 42-72
    • Summers, S.A.1
  • 270
    • 33847332202 scopus 로고    scopus 로고
    • Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturatedfat-, and obesity-induced insulin resistance
    • Holland WL, Brozinick JT, Wang LP, et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturatedfat-, and obesity-induced insulin resistance. Cell Metab. 2007;5:167-179.
    • (2007) Cell Metab. , vol.5 , pp. 167-179
    • Holland, W.L.1    Brozinick, J.T.2    Wang, L.P.3
  • 272
    • 58149484392 scopus 로고    scopus 로고
    • Inflammation and endoplasmic reticulum stress in obesity and diabetes
    • Hotamisligil GS. Inflammation and endoplasmic reticulum stress in obesity and diabetes. Int J Obes (Lond). 2008; 32(suppl 7):S52-S54.
    • (2008) Int J Obes (Lond). , vol.32 , Issue.SUPPL. 7
    • Hotamisligil, G.S.1
  • 273
    • 79151478555 scopus 로고    scopus 로고
    • Type 2 diabetes as an inflammatory disease
    • Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11:98-107.
    • (2011) Nat Rev Immunol. , vol.11 , pp. 98-107
    • Donath, M.Y.1    Shoelson, S.E.2
  • 274
    • 0037373449 scopus 로고    scopus 로고
    • Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males
    • Borras C, Sastre J, Garcia-Sala D, Lloret A, Pallardo FV, Vina J. Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males. Free Radic Biol Med. 2003;34:546-552.
    • (2003) Free Radic Biol Med. , vol.34 , pp. 546-552
    • Borras, C.1    Sastre, J.2    Garcia-Sala, D.3    Lloret, A.4    Pallardo, F.V.5    Vina, J.6
  • 275
    • 58149464712 scopus 로고    scopus 로고
    • Glutathione peroxidase 3 mediates the antioxidant effect of peroxisome proliferator-activated receptor γ in human skeletal muscle cells
    • Chung SS, Kim M, Youn BS, et al. Glutathione peroxidase 3 mediates the antioxidant effect of peroxisome proliferator-activated receptor γ in human skeletal muscle cells. Mol Cell Biol. 2009;29:20-30.
    • (2009) Mol Cell Biol. , vol.29 , pp. 20-30
    • Chung, S.S.1    Kim, M.2    Youn, B.S.3
  • 276
    • 63449132586 scopus 로고    scopus 로고
    • Validation of candidate causal genes for obesity that affect shared metabolic pathways and networks
    • Yang X, Deignan JL, Qi H, et al. Validation of candidate causal genes for obesity that affect shared metabolic pathways and networks. Nat Genet. 2009;41:415-423.
    • (2009) Nat Genet. , vol.41 , pp. 415-423
    • Yang, X.1    Deignan, J.L.2    Qi, H.3
  • 277
    • 0037477855 scopus 로고    scopus 로고
    • Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1
    • Patti ME, Butte AJ, Crunkhorn S, et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc Natl Acad Sci USA. 2003;100:8466-8471.
    • (2003) Proc Natl Acad Sci USA. , vol.100 , pp. 8466-8471
    • Patti, M.E.1    Butte, A.J.2    Crunkhorn, S.3
  • 278
    • 34248141686 scopus 로고    scopus 로고
    • Impaired mitochondrial substrate oxidation in muscle of insulin-resistant offspring of type 2 diabetic patients
    • Befroy DE, Petersen KF, Dufour S, et al. Impaired mitochondrial substrate oxidation in muscle of insulin-resistant offspring of type 2 diabetic patients. Diabetes. 2007;56: 1376-1381.
    • (2007) Diabetes. , vol.56 , pp. 1376-1381
    • Befroy, D.E.1    Petersen, K.F.2    Dufour, S.3
  • 279
    • 31044433308 scopus 로고    scopus 로고
    • Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents
    • Morino K, Petersen KF, Dufour S, et al. Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest. 2005;115:3587-3593.
    • (2005) J Clin Invest. , vol.115 , pp. 3587-3593
    • Morino, K.1    Petersen, K.F.2    Dufour, S.3
  • 280
    • 1642377274 scopus 로고    scopus 로고
    • Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes
    • Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med. 2004;350:664-671.
    • (2004) N Engl J Med. , vol.350 , pp. 664-671
    • Petersen, K.F.1    Dufour, S.2    Befroy, D.3    Garcia, R.4    Shulman, G.I.5
  • 281
    • 57649231744 scopus 로고    scopus 로고
    • Gender dimorphism in high-fat-diet-induced insulin resistance in skeletal muscle of aged rats
    • Gomez-Perez Y, Amengual-Cladera E, Catala-Niell A, et al. Gender dimorphism in high-fat-diet-induced insulin resistance in skeletal muscle of aged rats. Cell Physiol Biochem. 2008;22:539-548.
    • (2008) Cell Physiol Biochem. , vol.22 , pp. 539-548
    • Gomez-Perez, Y.1    Amengual-Cladera, E.2    Catala-Niell, A.3
  • 282
    • 0036066970 scopus 로고    scopus 로고
    • Knockout models are useful tools to dissect the pathophysiology and genetics of insulin resistance
    • Mauvais-Jarvis F, Kulkarni RN, Kahn CR. Knockout models are useful tools to dissect the pathophysiology and genetics of insulin resistance. Clin Endocrinol (Oxf). 2002; 57:1-9.
    • (2002) Clin Endocrinol (Oxf). , vol.57 , pp. 1-9
    • Mauvais-Jarvis, F.1    Kulkarni, R.N.2    Kahn, C.R.3
  • 283
    • 70349778705 scopus 로고    scopus 로고
    • Neuroprotective and anti-inflammatory effects of estrogen receptor ligand treatment in mice
    • Tiwari-Woodruff S, Voskuhl RR. Neuroprotective and anti-inflammatory effects of estrogen receptor ligand treatment in mice. J Neurol Sci. 2009;286:81-85.
    • (2009) J Neurol Sci. , vol.286 , pp. 81-85
    • Tiwari-Woodruff, S.1    Voskuhl, R.R.2
  • 284
    • 84857374216 scopus 로고    scopus 로고
    • 2011 Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration
    • Spence RD, Voskuhl RR. 2011 Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front Neuroendocrinol. 2012;33:105-115.
    • (2012) Front Neuroendocrinol. , vol.33 , pp. 105-115
    • Spence, R.D.1    Voskuhl, R.R.2
  • 285
    • 70349559752 scopus 로고    scopus 로고
    • Estrogen treatment decreases matrix metalloproteinase (MMP)-9 in autoimmune demyelinating disease through estrogen receptor α (ERα)
    • Gold SM, Sasidhar MV, Morales LB, et al. Estrogen treatment decreases matrix metalloproteinase (MMP)-9 in autoimmune demyelinating disease through estrogen receptor α (ERα). Lab Invest. 2009;89:1076-1083.
    • (2009) Lab Invest. , vol.89 , pp. 1076-1083
    • Gold, S.M.1    Sasidhar, M.V.2    Morales, L.B.3
  • 286
    • 0036167492 scopus 로고    scopus 로고
    • Pregnancy hormones prevent diabetes and reduce lymphocytic infiltration of islets in the NOD mouse
    • Atwater I, Gondos B, DiBartolomeo R, Bazaes R, Jovanovic L. Pregnancy hormones prevent diabetes and reduce lymphocytic infiltration of islets in the NOD mouse. Ann Clin Lab Sci. 2002;32:87-92.
    • (2002) Ann Clin Lab Sci. , vol.32 , pp. 87-92
    • Atwater, I.1    Gondos, B.2    DiBartolomeo, R.3    Bazaes, R.4    Jovanovic, L.5
  • 287
    • 77952497806 scopus 로고    scopus 로고
    • Suppression of the inflammatory response in experimental arthritis is mediated via estrogen receptor α but not estrogen receptor β
    • Dulos J, Vijn P, van Doorn C, et al. Suppression of the inflammatory response in experimental arthritis is mediated via estrogen receptor α but not estrogen receptor β. Arthritis Res Ther. 2010;12:R101.
    • (2010) Arthritis Res Ther. , vol.12
    • Dulos, J.1    Vijn, P.2    van Doorn, C.3
  • 288
    • 77950539083 scopus 로고    scopus 로고
    • Endogenous estrogen regulation of inflammatory arthritis and cytokine expression in male mice, predominantly via estrogen receptor α
    • Yang YH, Ngo D, Jones M, Simpson E, Fritzemeier KH, Morand EF. Endogenous estrogen regulation of inflammatory arthritis and cytokine expression in male mice, predominantly via estrogen receptor α. Arthritis Rheum. 2010;62:1017-1025.
    • (2010) Arthritis Rheum. , vol.62 , pp. 1017-1025
    • Yang, Y.H.1    Ngo, D.2    Jones, M.3    Simpson, E.4    Fritzemeier, K.H.5    Morand, E.F.6
  • 289
    • 57649205323 scopus 로고    scopus 로고
    • Estrogen and progesterone affect responses to malaria infection in female C57BL/6 mice
    • Klein PW, Easterbrook JD, Lalime EN, Klein SL. Estrogen and progesterone affect responses to malaria infection in female C57BL/6 mice. Gend Med. 2008;5:423-433.
    • (2008) Gend Med. , vol.5 , pp. 423-433
    • Klein, P.W.1    Easterbrook, J.D.2    Lalime, E.N.3    Klein, S.L.4
  • 290
    • 73649112085 scopus 로고    scopus 로고
    • Estrogen receptor-α as a drug target candidate for preventing lung inflammation
    • Vegeto E, Cuzzocrea S, Crisafulli C, et al. Estrogen receptor-α as a drug target candidate for preventing lung inflammation. Endocrinology. 2010;151:174-184.
    • (2010) Endocrinology. , vol.151 , pp. 174-184
    • Vegeto, E.1    Cuzzocrea, S.2    Crisafulli, C.3
  • 291
    • 77955485472 scopus 로고    scopus 로고
    • 17β-Estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor α signaling in macrophages in vivo
    • Calippe B, Douin-Echinard V, Delpy L, et al.17β-Estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor α signaling in macrophages in vivo. J Immunol. 2010;185:1169-1176.
    • (2010) J Immunol. , vol.185 , pp. 1169-1176
    • Calippe, B.1    Douin-Echinard, V.2    Delpy, L.3
  • 292
    • 75149196834 scopus 로고    scopus 로고
    • The role of sex hormones in the development of Th2 immunity in a gender-biased model of Trichuris muris infection
    • Hepworth MR, Hardman MJ, Grencis RK. The role of sex hormones in the development of Th2 immunity in a gender-biased model of Trichuris muris infection. Eur J Immunol. 2010;40:406-416.
    • (2010) Eur J Immunol. , vol.40 , pp. 406-416
    • Hepworth, M.R.1    Hardman, M.J.2    Grencis, R.K.3
  • 294
    • 79959825685 scopus 로고    scopus 로고
    • Estradiol administration controls eosinophilia through estrogen receptor-α activation during acute peritoneal inflammation
    • Douin-Echinard V, Calippe B, Billon-Gales A, et al. Estradiol administration controls eosinophilia through estrogen receptor-α activation during acute peritoneal inflammation. J Leukoc Biol. 2011;90:145-154.
    • (2011) J Leukoc Biol. , vol.90 , pp. 145-154
    • Douin-Echinard, V.1    Calippe, B.2    Billon-Gales, A.3
  • 295
    • 77951918926 scopus 로고    scopus 로고
    • Macrophages, inflammation, and insulin resistance
    • Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219-246.
    • (2010) Annu Rev Physiol. , vol.72 , pp. 219-246
    • Olefsky, J.M.1    Glass, C.K.2
  • 296
    • 80355133234 scopus 로고    scopus 로고
    • Macrophage-mediated inflammation in metabolic disease
    • Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 2011;11:738-749.
    • (2011) Nat Rev Immunol. , vol.11 , pp. 738-749
    • Chawla, A.1    Nguyen, K.D.2    Goh, Y.P.3
  • 297
    • 80053630034 scopus 로고    scopus 로고
    • Myeloid-specific estrogen receptor α deficiency impairs metabolic homeostasis and accelerates atherosclerotic lesion development
    • Ribas V, Drew BG, Le JA, et al. Myeloid-specific estrogen receptor α deficiency impairs metabolic homeostasis and accelerates atherosclerotic lesion development. Proc Natl Acad Sci USA. 2011;108:16457-16462.
    • (2011) Proc Natl Acad Sci USA. , vol.108 , pp. 16457-16462
    • Ribas, V.1    Drew, B.G.2    Le, J.A.3
  • 298
    • 77955606282 scopus 로고    scopus 로고
    • Extranuclear estrogen receptor-α stimulates NeuroD1 binding to the insulin promoter and favors insulin synthesis
    • Wong WP, Tiano JP, Liu S, et al. Extranuclear estrogen receptor-α stimulates NeuroD1 binding to the insulin promoter and favors insulin synthesis. Proc Natl Acad Sci USA. 2010;107:13057-13062.
    • (2010) Proc Natl Acad Sci USA. , vol.107 , pp. 13057-13062
    • Wong, W.P.1    Tiano, J.P.2    Liu, S.3
  • 299
    • 79960977229 scopus 로고    scopus 로고
    • Estrogen receptor activation reduces lipid synthesis in pancreatic islets and prevents β cell failure in rodent models of type 2 diabetes
    • Tiano JP, Delghingaro-Augusto V, Le May C, et al. Estrogen receptor activation reduces lipid synthesis in pancreatic islets and prevents β cell failure in rodent models of type 2 diabetes. J Clin Invest. 2011;121:3331-3342.
    • (2011) J Clin Invest. , vol.121 , pp. 3331-3342
    • Tiano, J.P.1    Delghingaro-Augusto, V.2    Le May, C.3
  • 300
    • 84862219400 scopus 로고    scopus 로고
    • Selective estrogen receptor modulation in pancreaticβ-cells and the prevention of type 2 diabetes
    • Tiano J, Mauvais-Jarvis F. Selective estrogen receptor modulation in pancreaticβ-cells and the prevention of type 2 diabetes. Islets. 2012;4:173-176.
    • (2012) Islets. , vol.4 , pp. 173-176
    • Tiano, J.1    Mauvais-Jarvis, F.2
  • 301
    • 84862737646 scopus 로고    scopus 로고
    • Molecular mechanisms of estrogen receptors' suppression of lipogenesis in pancreatic β-cells
    • Tiano JP, Mauvais-Jarvis F. Molecular mechanisms of estrogen receptors' suppression of lipogenesis in pancreatic β-cells. Endocrinology. 2012;153:2997-3005.
    • (2012) Endocrinology. , vol.153 , pp. 2997-3005
    • Tiano, J.P.1    Mauvais-Jarvis, F.2
  • 302
    • 44349121098 scopus 로고    scopus 로고
    • Pancreatic insulin content regulation by the estrogen receptor ER α
    • Alonso-Magdalena P, Ropero AB, Carrera MP, et al. Pancreatic insulin content regulation by the estrogen receptor ER α. PLoS One. 2008;3:e2069.
    • (2008) PLoS One. , vol.3
    • Alonso-Magdalena, P.1    Ropero, A.B.2    Carrera, M.P.3
  • 303
    • 84857508749 scopus 로고    scopus 로고
    • The 2012 hormone therapy position statement of: The North American Menopause Society.
    • The 2012 hormone therapy position statement of: The North American Menopause Society. Menopause. 2012; 19:257-271.
    • (2012) Menopause , vol.19 , pp. 257-271
  • 304
    • 34249682775 scopus 로고    scopus 로고
    • Chronic activation of liver X receptor induces β-cell apoptosis through hyperactivation of lipogenesis: liver X receptor-mediated lipotoxicity in pancreatic β-cells
    • Choe SS, Choi AH, Lee JW, et al. Chronic activation of liver X receptor induces β-cell apoptosis through hyperactivation of lipogenesis: liver X receptor-mediated lipotoxicity in pancreatic β-cells. Diabetes. 2007;56:1534-1543.
    • (2007) Diabetes. , vol.56 , pp. 1534-1543
    • Choe, S.S.1    Choi, A.H.2    Lee, J.W.3
  • 305
    • 33745124662 scopus 로고    scopus 로고
    • Estrogens protect pancreatic β-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice
    • Le May C, Chu K, Hu M, et al. Estrogens protect pancreatic β-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. Proc Natl Acad Sci USA. 2006; 103:9232-9237.
    • (2006) Proc Natl Acad Sci USA. , vol.103 , pp. 9232-9237
    • Le May, C.1    Chu, K.2    Hu, M.3
  • 306
    • 70349649964 scopus 로고    scopus 로고
    • Importance of extranuclear estrogen receptor-α and membraneGprotein-coupled estrogen receptor in pancreatic islet survival
    • Liu S, LeMayC,WongWP,et al. Importance of extranuclear estrogen receptor-α and membraneGprotein-coupled estrogen receptor in pancreatic islet survival. Diabetes. 2009;58: 2292-2302.
    • (2009) Diabetes. , vol.58 , pp. 2292-2302
    • Liu, S.1    LeMay, C.2    Wong, W.P.3
  • 307
    • 77149168602 scopus 로고    scopus 로고
    • Rapid, nongenomic estrogen actions protect pancreatic islet survival
    • Liu S, Mauvais-Jarvis F. Rapid, nongenomic estrogen actions protect pancreatic islet survival. Islets. 2009;1:273-275.
    • (2009) Islets. , vol.1 , pp. 273-275
    • Liu, S.1    Mauvais-Jarvis, F.2
  • 308
    • 79959781820 scopus 로고    scopus 로고
    • The liver receptor homolog-1 (LRH-1) is expressed in human islets and protects β-cells against stress-induced apoptosis
    • BaquieM,St-Onge L, Kerr-Conte J, et al. The liver receptor homolog-1 (LRH-1) is expressed in human islets and protects β-cells against stress-induced apoptosis. Hum Mol Genet. 2011;20:2823-2833.
    • (2011) Hum Mol Genet. , vol.20 , pp. 2823-2833
    • Baquie, M.1    St-Onge, L.2    Kerr-Conte, J.3
  • 309
    • 73249129499 scopus 로고    scopus 로고
    • Rapid regulation of K(ATP) channel activity by 17β-estradiol in pancreatic β-cells involves the estrogen receptor β and the atrial natriuretic peptide receptor
    • Soriano S, Ropero AB, Alonso-Magdalena P, et al. Rapid regulation of K(ATP) channel activity by 17β-estradiol in pancreatic β-cells involves the estrogen receptor β and the atrial natriuretic peptide receptor. Mol Endocrinol. 2009; 23:1973-1982.
    • (2009) Mol Endocrinol. , vol.23 , pp. 1973-1982
    • Soriano, S.1    Ropero, A.B.2    Alonso-Magdalena, P.3
  • 310
    • 84856762350 scopus 로고    scopus 로고
    • Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: role of estrogen receptor β
    • Soriano S, Alonso-Magdalena P, Garcia-Arevalo M, et al. Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: role of estrogen receptor β. PLoS One. 2012;7:e31109.
    • (2012) PLoS One. , vol.7
    • Soriano, S.1    Alonso-Magdalena, P.2    Garcia-Arevalo, M.3
  • 311
    • 84862219400 scopus 로고    scopus 로고
    • Selective estrogen receptor modulation in pancreaticβ-cells and the prevention of type 2 diabetes
    • Tiano J, Mauvais-Jarvis F. Selective estrogen receptor modulation in pancreaticβ-cells and the prevention of type 2 diabetes. Islets. 2012;4:173-176.
    • (2012) Islets. , vol.4 , pp. 173-176
    • Tiano, J.1    Mauvais-Jarvis, F.2
  • 312
    • 77949566651 scopus 로고    scopus 로고
    • Activation of G protein-coupled receptor 30 modulates hormone secretion and counteracts cytokine-induced apoptosis in pancreatic islets of female mice
    • Balhuizen A, Kumar R, Amisten S, Lundquist I, Salehi A. Activation of G protein-coupled receptor 30 modulates hormone secretion and counteracts cytokine-induced apoptosis in pancreatic islets of female mice. Mol Cell Endocrinol. 2010;320:16-24.
    • (2010) Mol Cell Endocrinol. , vol.320 , pp. 16-24
    • Balhuizen, A.1    Kumar, R.2    Amisten, S.3    Lundquist, I.4    Salehi, A.5
  • 313
    • 79959457594 scopus 로고    scopus 로고
    • Insulinotropic and antidiabetic effects of 17β-estradiol and the GPR30 agonist G-1 on human pancreatic islets
    • Kumar R, Balhuizen A, Amisten S, Lundquist I, Salehi A. Insulinotropic and antidiabetic effects of 17β-estradiol and the GPR30 agonist G-1 on human pancreatic islets. Endocrinology. 2011;152:2568-2579.
    • (2011) Endocrinology. , vol.152 , pp. 2568-2579
    • Kumar, R.1    Balhuizen, A.2    Amisten, S.3    Lundquist, I.4    Salehi, A.5
  • 314
    • 79960705272 scopus 로고    scopus 로고
    • Mechanisms of estradiol-induced insulin secretion by the G protein-coupled estrogen receptor GPR30/GPER in pancreatic β-cells
    • Sharma G, Prossnitz ER. Mechanisms of estradiol-induced insulin secretion by the G protein-coupled estrogen receptor GPR30/GPER in pancreatic β-cells. Endocrinology. 2011;152:3030-3039.
    • (2011) Endocrinology. , vol.152 , pp. 3030-3039
    • Sharma, G.1    Prossnitz, E.R.2
  • 315
    • 0037110453 scopus 로고    scopus 로고
    • 17β-Estradiol protects isolated human pancreatic islets against proinflammatory cytokineinduced cell death: molecular mechanisms and islet functionality
    • Contreras JL, Smyth CA, Bilbao G, Young CJ, Thompson JA, Eckhoff DE. 17β-Estradiol protects isolated human pancreatic islets against proinflammatory cytokineinduced cell death: molecular mechanisms and islet functionality. Transplantation. 2002;74:1252-1259.
    • (2002) Transplantation. , vol.74 , pp. 1252-1259
    • Contreras, J.L.1    Smyth, C.A.2    Bilbao, G.3    Young, C.J.4    Thompson, J.A.5    Eckhoff, D.E.6
  • 316
    • 33646357008 scopus 로고    scopus 로고
    • Sexual function and endocrine profile in fertile women with type 1 diabetes
    • Salonia A, Lanzi R, Scavini M, et al. Sexual function and endocrine profile in fertile women with type 1 diabetes. Diabetes Care. 2006;29:312-316.
    • (2006) Diabetes Care. , vol.29 , pp. 312-316
    • Salonia, A.1    Lanzi, R.2    Scavini, M.3
  • 317
    • 84878659317 scopus 로고    scopus 로고
    • Oestrogens improve human pancreatic islet transplantation in a mouse model of insulin deficient diabetes
    • Liu S, Kilic G, Meyers MS, et al. Oestrogens improve human pancreatic islet transplantation in a mouse model of insulin deficient diabetes. Diabetologia. 2013;56:370-381.
    • (2013) Diabetologia. , vol.56 , pp. 370-381
    • Liu, S.1    Kilic, G.2    Meyers, M.S.3
  • 318
    • 84870902173 scopus 로고    scopus 로고
    • Targeted estrogen delivery reverses the metabolic syndrome
    • Finan B, Yang B, Ottaway N, et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat Med. 2012; 18:1847-1856.
    • (2012) Nat Med. , vol.18 , pp. 1847-1856
    • Finan, B.1    Yang, B.2    Ottaway, N.3
  • 319
    • 84892627122 scopus 로고    scopus 로고
    • A glucagon-like peptide-1-estrogen fusion peptide shows en-hanced efficacy in preventing insulin-deficient diabetes in mice
    • Tiano J, Finan B, DiMarchi R, Mauvais-Jarvis F. A glucagon-like peptide-1-estrogen fusion peptide shows en-hanced efficacy in preventing insulin-deficient diabetes in mice. Endocr Rev. 2012;33:OR21-OR26.
    • (2012) Endocr Rev. , vol.33
    • Tiano, J.1    Finan, B.2    DiMarchi, R.3    Mauvais-Jarvis, F.4
  • 320
    • 54349083290 scopus 로고    scopus 로고
    • Gender-specific expression and mechanism of regulation of estrogen sulfotransferase in adipose tissues of the mouse
    • Khor VK, Tong MH, Qian Y, Song WC. Gender-specific expression and mechanism of regulation of estrogen sulfotransferase in adipose tissues of the mouse. Endocrinology. 2008;149:5440-5448.
    • (2008) Endocrinology. , vol.149 , pp. 5440-5448
    • Khor, V.K.1    Tong, M.H.2    Qian, Y.3    Song, W.C.4
  • 322
    • 79960086104 scopus 로고    scopus 로고
    • Estrogen sulfotransferase is expressed in subcutaneous adipose tissue of obese humans in association with TNF-α and SOCS3
    • Ahima RS, Stanley TL, Khor VK, Zanni MV, Grinspoon SK. Estrogen sulfotransferase is expressed in subcutaneous adipose tissue of obese humans in association with TNF-α and SOCS3. J Clin Endocrinol Metab. 2011;96:E1153-E1158.
    • (2011) J Clin Endocrinol Metab. , vol.96
    • Ahima, R.S.1    Stanley, T.L.2    Khor, V.K.3    Zanni, M.V.4    Grinspoon, S.K.5
  • 323
    • 0028268953 scopus 로고
    • Obesity-induced diabetes (diabesity) in C57BL/KsJ mice produces aberrant transregulation of sex steroid sulfotransferase genes
    • Leiter EH, Chapman HD. Obesity-induced diabetes (diabesity) in C57BL/KsJ mice produces aberrant transregulation of sex steroid sulfotransferase genes. J Clin Invest. 1994;93:2007-2013.
    • (1994) J Clin Invest. , vol.93 , pp. 2007-2013
    • Leiter, E.H.1    Chapman, H.D.2
  • 324
    • 84861867457 scopus 로고    scopus 로고
    • Sex-specific effect of estrogen sulfotransferase on mouse models of type 2 diabetes
    • Gao J, He J, Shi X, et al. Sex-specific effect of estrogen sulfotransferase on mouse models of type 2 diabetes. Diabetes. 2012;61:1543-1551.
    • (2012) Diabetes. , vol.61 , pp. 1543-1551
    • Gao, J.1    He, J.2    Shi, X.3
  • 326
    • 0033102731 scopus 로고    scopus 로고
    • Comparison of Alora estradiol matrix transdermal delivery system with oral conjugated equine estrogen therapy in relieving menopausal symptoms
    • 326. Good WR, John VA, Ramirez M, Higgins JE. Comparison of Alora estradiol matrix transdermal delivery system with oral conjugated equine estrogen therapy in relieving menopausal symptoms. Alora Study Group. Climacteric. 1999; 2:29-36.
    • (1999) Alora Study Group. Climacteric. , vol.2 , pp. 29-36
    • Good, W.R.1    John, V.A.2    Ramirez, M.3    Higgins, J.E.4
  • 328
    • 0019975473 scopus 로고
    • Effects of percutaneous estradiol and conjugated estrogens on the level of plasma proteins and triglycerides in postmenopausal women
    • Elkik F, Gompel A, Mercier-Bodard C, et al. Effects of percutaneous estradiol and conjugated estrogens on the level of plasma proteins and triglycerides in postmenopausal women. Am J Obstet Gynecol. 1982;143:888-892.
    • (1982) Am J Obstet Gynecol. , vol.143 , pp. 888-892
    • Elkik, F.1    Gompel, A.2    Mercier-Bodard, C.3
  • 329
    • 33751160143 scopus 로고    scopus 로고
    • comparison of oral and transdermal short-term estrogen therapy in postmenopausal women with metabolic syndrome
    • ChuMC,Cosper P, Nakhuda GS, Lobo RA.Acomparison of oral and transdermal short-term estrogen therapy in postmenopausal women with metabolic syndrome. Fertil Steril. 2006;86:1669-1675.
    • (2006) Fertil Steril. , vol.86 , pp. 1669-1675
    • Chu, M.C.1    Cosper, P.2    Nakhuda, G.S.3    Lobo, R.A.A.4
  • 330
    • 54849427908 scopus 로고    scopus 로고
    • Metabolic syndrome in postmenopausal women: the influence of oral or transdermal estradiol on inflammation and coagulation markers
    • Chu MC, Cushman M, Solomon R, Lobo RA. Metabolic syndrome in postmenopausal women: the influence of oral or transdermal estradiol on inflammation and coagulation markers.AmJ Obstet Gynecol. 2008;199:526.e1-526.e7.
    • (2008) AmJ Obstet Gynecol. , vol.199
    • Chu, M.C.1    Cushman, M.2    Solomon, R.3    Lobo, R.A.4
  • 331
    • 33746862148 scopus 로고    scopus 로고
    • Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women
    • Salpeter SR, Walsh JM, OrmistonTM,Greyber E, Buckley NS, Salpeter EE. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab. 2006;8: 538-554.
    • (2006) Diabetes Obes Metab. , vol.8 , pp. 538-554
    • Salpeter, S.R.1    Walsh, J.M.2    Ormiston, T.M.3    Greyber, E.4    Buckley, N.S.5    Salpeter, E.E.6
  • 332
    • 0034687825 scopus 로고    scopus 로고
    • A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease
    • Grodstein F, Manson JE, Colditz GA, Willett WC, Speizer FE, Stampfer MJ. A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease. Ann Intern Med. 2000;133: 933-941.
    • (2000) Ann Intern Med. , vol.133 , pp. 933-941
    • Grodstein, F.1    Manson, J.E.2    Colditz, G.A.3    Willett, W.C.4    Speizer, F.E.5    Stampfer, M.J.6
  • 333
    • 71149117152 scopus 로고    scopus 로고
    • Bayesian meta-analysis of hormone therapy and mortality in younger postmenopausal women
    • Salpeter SR, Cheng J, Thabane L, Buckley NS, Salpeter EE. Bayesian meta-analysis of hormone therapy and mortality in younger postmenopausal women.AmJ Med. 2009;122: 1016-1022.e1.
    • (2009) AmJ Med. , vol.122
    • Salpeter, S.R.1    Cheng, J.2    Thabane, L.3    Buckley, N.S.4    Salpeter, E.E.5
  • 334
    • 0032547326 scopus 로고    scopus 로고
    • Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women
    • Hulley S, Grady D, Bush T, et al. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA. 1998;280:605-613.
    • (1998) Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA. , vol.280 , pp. 605-613
    • Hulley, S.1    Grady, D.2    Bush, T.3
  • 336
    • 1842867053 scopus 로고    scopus 로고
    • Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial
    • Anderson GL, Limacher M, Assaf AR, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA. 2004;291:1701-1712.
    • (2004) JAMA. , vol.291 , pp. 1701-1712
    • Anderson, G.L.1    Limacher, M.2    Assaf, A.R.3
  • 337
  • 338
    • 0036204302 scopus 로고    scopus 로고
    • Tamoxifen therapy and hepatic steatosis
    • Coskun U, Toruner FB, Gunel N. Tamoxifen therapy and hepatic steatosis. Neoplasma. 2002;49:61-64.
    • (2002) Neoplasma. , vol.49 , pp. 61-64
    • Coskun, U.1    Toruner, F.B.2    Gunel, N.3
  • 339
    • 0037216370 scopus 로고    scopus 로고
    • Effects of tamoxifen on hepatic fat content and the development of hepatic steatosis in patients with breast cancer: high frequency of involvement and rapid reversal after completion of tamoxifen therapy
    • NishinoM,HayakawaK,NakamuraY,MorimotoT,Mukaihara S. Effects of tamoxifen on hepatic fat content and the development of hepatic steatosis in patients with breast cancer: high frequency of involvement and rapid reversal after completion of tamoxifen therapy. AJR Am J Roentgenol. 2003;180:129-134.
    • (2003) AJR Am J Roentgenol. , vol.180 , pp. 129-134
    • Nishino, M.1    Hayakawa, K.2    Nakamura, Y.3    Morimoto, T.4    Mukaihara, S.5
  • 340
    • 21744460524 scopus 로고    scopus 로고
    • Transcript and metabolite analysis of the effects of tamoxifen in rat liver reveals inhibition of fatty acid synthesis in the presence of hepatic steatosis
    • Lelliott CJ, Lopez M, Curtis RK, et al. Transcript and metabolite analysis of the effects of tamoxifen in rat liver reveals inhibition of fatty acid synthesis in the presence of hepatic steatosis. FASEB J. 2005;19:1108-1119.
    • (2005) FASEB J. , vol.19 , pp. 1108-1119
    • Lelliott, C.J.1    Lopez, M.2    Curtis, R.K.3
  • 341
    • 33845593130 scopus 로고    scopus 로고
    • Causes and prevention of tamoxifen-induced accumulation of triacylglycerol in rat liver
    • Gudbrandsen OA, Rost TH, Berge RK. Causes and prevention of tamoxifen-induced accumulation of triacylglycerol in rat liver. J Lipid Res. 2006;47:2223-2232.
    • (2006) J Lipid Res. , vol.47 , pp. 2223-2232
    • Gudbrandsen, O.A.1    Rost, T.H.2    Berge, R.K.3
  • 342
    • 77957961658 scopus 로고    scopus 로고
    • Tamoxifen induces triacylglycerol accumulation in the mouse liver by activation of fatty acid synthesis
    • Cole LK, Jacobs RL, Vance DE. Tamoxifen induces triacylglycerol accumulation in the mouse liver by activation of fatty acid synthesis. Hepatology. 2010;52:1258-1265.
    • (2010) Hepatology. , vol.52 , pp. 1258-1265
    • Cole, L.K.1    Jacobs, R.L.2    Vance, D.E.3
  • 343
    • 33745294817 scopus 로고    scopus 로고
    • Tamoxifen-induced anorexia is associated with fatty acid synthase inhibition in the ventromedial nucleus of the hypothalamus and accumulation of malonyl-CoA
    • Lopez M, Lelliott CJ, Tovar S, et al. Tamoxifen-induced anorexia is associated with fatty acid synthase inhibition in the ventromedial nucleus of the hypothalamus and accumulation of malonyl-CoA. Diabetes. 2006;55:1327-1336.
    • (2006) Diabetes. , vol.55 , pp. 1327-1336
    • Lopez, M.1    Lelliott, C.J.2    Tovar, S.3
  • 344
    • 84860652546 scopus 로고    scopus 로고
    • Association between tamoxifen treatment and diabetes: a populationbased study
    • Lipscombe LL, Fischer HD, Yun L, et al. Association between tamoxifen treatment and diabetes: a populationbased study. Cancer. 2012;118:2615-2622.
    • (2012) Cancer. , vol.118 , pp. 2615-2622
    • Lipscombe, L.L.1    Fischer, H.D.2    Yun, L.3
  • 345
    • 0033581212 scopus 로고    scopus 로고
    • Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial
    • Ettinger B, Black DM, Mitlak BH, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA. 1999;282: 637-645.
    • (1999) Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA. , vol.282 , pp. 637-645
    • Ettinger, B.1    Black, D.M.2    Mitlak, B.H.3
  • 346
    • 3042543880 scopus 로고    scopus 로고
    • Estrogen and raloxifene modulate leptin and its receptor in hypothalamus and adipose tissue from ovariectomized rats
    • Meli R, Pacilio M, RasoGM,et al. Estrogen and raloxifene modulate leptin and its receptor in hypothalamus and adipose tissue from ovariectomized rats. Endocrinology. 2004;145:3115-3121.
    • (2004) Endocrinology. , vol.145 , pp. 3115-3121
    • Meli, R.1    Pacilio, M.2    Raso, G.M.3
  • 347
    • 33746045998 scopus 로고    scopus 로고
    • Serum leptin levels and body composition in postmenopausal women treated with tibolone and raloxifene
    • Tommaselli GA, Di Carlo C, Di Spiezio Sardo A, et al. Serum leptin levels and body composition in postmenopausal women treated with tibolone and raloxifene. Menopause. 2006;13:660-668.
    • (2006) Menopause. , vol.13 , pp. 660-668
    • Tommaselli, G.A.1    Di Carlo, C.2    Di Spiezio Sardo, A.3
  • 348
  • 349
    • 0037365104 scopus 로고    scopus 로고
    • Post hoc analysis of data from the Multiple Outcomes of Raloxifene Evaluation (MORE) trial on the effects of three years of raloxifene treatment on glycemic control and cardiovascular disease risk factors in women with and without type 2 diabetes
    • Barrett-Connor E, Ensrud KE, Harper K, et al. Post hoc analysis of data from the Multiple Outcomes of Raloxifene Evaluation (MORE) trial on the effects of three years of raloxifene treatment on glycemic control and cardiovascular disease risk factors in women with and without type 2 diabetes. Clin Ther. 2003;25:919-930.
    • (2003) Clin Ther. , vol.25 , pp. 919-930
    • Barrett-Connor, E.1    Ensrud, K.E.2    Harper, K.3
  • 350
    • 75149182867 scopus 로고    scopus 로고
    • Raloxifene and body composition and muscle strength in postmenopausal women: a randomized, double-blind, placebo-controlled trial
    • Jacobsen DE, Samson MM, Emmelot-Vonk MH, Verhaar HJ. Raloxifene and body composition and muscle strength in postmenopausal women: a randomized, double-blind, placebo-controlled trial. Eur J Endocrinol. 2010;162:371-376. 351. Andersson B, Johannsson G, Holm G, et al. Raloxifene does not affect insulin sensitivity or glycemic control in postmenopausal women with type 2 diabetes mellitus: a randomized clinical trial. J Clin Endocrinol Metab. 2002; 87:122-128.
    • (2010) Eur J Endocrinol. , vol.162 , pp. 371-376
    • Jacobsen, D.E.1    Samson, M.M.2    Emmelot-Vonk, M.H.3    Verhaar, H.J.4
  • 351
    • 0036148237 scopus 로고    scopus 로고
    • Raloxifene does not affect insulin sensitivity or glycemic control in postmenopausal women with type 2 diabetes mellitus: a randomized clinical trial
    • Andersson B, Johannsson G, Holm G, et al. Raloxifene does not affect insulin sensitivity or glycemic control in postmenopausal women with type 2 diabetes mellitus: a randomized clinical trial. J Clin Endocrinol Metab. 2002; 87:122-128.
    • (2002) J Clin Endocrinol Metab. , vol.87 , pp. 122-128
    • Andersson, B.1    Johannsson, G.2    Holm, G.3
  • 352
    • 40249093057 scopus 로고    scopus 로고
    • Effects of raloxifene on insulin sensitivity, β-cell function, and hepatic insulin extraction in normal postmenopausal women
    • Nagamani M, Szymajda A, Sepilian V, Urban RJ, Gilkison C. Effects of raloxifene on insulin sensitivity, β-cell function, and hepatic insulin extraction in normal postmenopausal women. Fertil Steril. 2008;89:614-619.
    • (2008) Fertil Steril. , vol.89 , pp. 614-619
    • Nagamani, M.1    Szymajda, A.2    Sepilian, V.3    Urban, R.J.4    Gilkison, C.5
  • 353
    • 21544472890 scopus 로고    scopus 로고
    • Effect of raloxifene on serum triglycerides in women with a history of hypertriglyceridemia while on oral estrogen therapy
    • Carr MC, Knopp RH, Brunzell JD, et al. Effect of raloxifene on serum triglycerides in women with a history of hypertriglyceridemia while on oral estrogen therapy. Diabetes Care. 2005;28:1555-1561.
    • (2005) Diabetes Care. , vol.28 , pp. 1555-1561
    • Carr, M.C.1    Knopp, R.H.2    Brunzell, J.D.3
  • 354
    • 56549102911 scopus 로고    scopus 로고
    • The pairing of a selective estrogen receptor modulator, bazedoxifene, with conjugated estrogens as a new paradigm for the treatment of menopausal symptoms and osteoporosis prevention
    • Kharode Y, Bodine PV, Miller CP, Lyttle CR, Komm BS. The pairing of a selective estrogen receptor modulator, bazedoxifene, with conjugated estrogens as a new paradigm for the treatment of menopausal symptoms and osteoporosis prevention. Endocrinology. 2008;149:6084-6091.
    • (2008) Endocrinology. , vol.149 , pp. 6084-6091
    • Kharode, Y.1    Bodine, P.V.2    Miller, C.P.3    Lyttle, C.R.4    Komm, B.S.5
  • 355
    • 57849153536 scopus 로고    scopus 로고
    • A new approach to menopausal therapy: the tissue selective estrogen complex
    • Komm BS. A new approach to menopausal therapy: the tissue selective estrogen complex. Reprod Sci. 2008;15: 984-992.
    • (2008) Reprod Sci. , vol.15 , pp. 984-992
    • Komm, B.S.1
  • 356
    • 84859462129 scopus 로고    scopus 로고
    • Selective estrogen receptor modulator promotes weight loss in ovariectomized female rhesus monkeys (Macaca mulatta) by decreasing food intake and increasing activity
    • Sullivan EL, Shearin J, Koegler FH, Cameron JL. Selective estrogen receptor modulator promotes weight loss in ovariectomized female rhesus monkeys (Macaca mulatta) by decreasing food intake and increasing activity. Am J Physiol Endocrinol Metab. 2012;302:E759-E767.
    • (2012) Am J Physiol Endocrinol Metab. , vol.302
    • Sullivan, E.L.1    Shearin, J.2    Koegler, F.H.3    Cameron, J.L.4
  • 357
    • 0038210242 scopus 로고    scopus 로고
    • Aromatase inhibitors in breast cancer
    • Smith IE, Dowsett M. Aromatase inhibitors in breast cancer. N Engl J Med. 2003;348:2431-2442.
    • (2003) N Engl J Med. , vol.348 , pp. 2431-2442
    • Smith, I.E.1    Dowsett, M.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.