47
Views
0
CrossRef citations to date
0
Altmetric
Review

Perinatal androgen exposure and adipose tissue programming: is there an impact on body weight fate?

, &

References

  • WHO. Obesity and overweight. WHO; 2013
  • Gluckman PD, Hanson MA. The developmental origins of the metabolic syndrome. Trends Endocrinol Metab 2004;15:183-7
  • Rogers I; Group E-BS. The influence of birthweight and intrauterine environment on adiposity and fat distribution in later life. Int J Obes Relat Metab Disord 2003;27:755-77
  • Vo T, Hardy DB. Molecular mechanisms underlying the fetal programming of adult disease. J Cell Commun Signal 2012;6:139-53
  • Zambrano E, Guzman C, Rodriguez-Gonzalez GL, et al. Fetal programming of sexual development and reproductive function. Mol Cell Endocrinol 2014;382:538-49
  • Burdge GC, Hanson MA, Slater-Jefferies JL, et al. Epigenetic regulation of transcription: a mechanism for inducing variations in phenotype (fetal programming) by differences in nutrition during early life? Br J Nutr 2007;97:1036-46
  • O’Sullivan L, Combes AN, Moritz KM. Epigenetics and developmental programming of adult onset diseases. Pediatr Nephrol 2012;27:2175-82
  • Ugele B, St-Pierre MV, Pihusch M, et al. Characterization and identification of steroid sulfate transporters of human placenta. Am J Physiol Endocrinol Metab 2003;284:E390-8
  • Li Y, Isomaa V, Pulkka A, et al. Expression of 3beta-hydroxysteroid dehydrogenase type 1, P450 aromatase, and 17beta-hydroxysteroid dehydrogenase types 1, 2, 5 and 7 mRNAs in human early and mid-gestation placentas. Placenta 2005;26:387-92
  • Pasqualini JR. Enzymes involved in the formation and transformation of steroid hormones in the fetal and placental compartments. J Steroid Biochem Mol Biol 2005;97:401-15
  • Meloche CA, Falany CN. Expression and characterization of the human 3 beta-hydroxysteroid sulfotransferases (SULT2B1a and SULT2B1b). J Steroid Biochem Mol Biol 2001;77:261-9
  • Geese WJ, Raftogianis RB. Biochemical characterization and tissue distribution of human SULT2B1. Biochem Biophys Res Commun 2001;288:280-9
  • Mitra P, Audus KL. Expression and functional activities of selected sulfotransferase isoforms in BeWo cells and primary cytotrophoblast cells. Biochem Pharmacol 2009;78:1475-82
  • Tagawa N, Hidaka Y, Takano T, et al. Serum concentrations of dehydroepiandrosterone and dehydroepiandrosterone sulfate and their relation to cytokine production during and after normal pregnancy. Clin Chim Acta 2004;340:187-93
  • Morisset AS, Dube MC, Drolet R, et al. Androgens in the maternal and fetal circulation: association with insulin resistance. J Matern-Fetal Neo M 2013;26:513-19
  • Kuijper EAM, Ket JCF, Caanen MR, et al. Reproductive hormone concentrations in pregnancy and neonates: a systematic review. Reprod Biomed Online 2013;27:33-63
  • Kerlan V, Nahoul K, Lemartelot MT, et al. Longitudinal-Study of Maternal Plasma Bioavailable Testosterone and Androstanediol Glucuronide Levels during Pregnancy. Clin Endocrinol 1994;40:263-7
  • van de Beek C, Thijssen JHH, Cohen-Kettenis PT, et al. Relationships between sex hormones assessed in amniotic fluid, and maternal and umbilical cord serum: What is the best source of information to investigate the effects of fetal hormonal exposure? Horm Behav 2004;46:663-9
  • Rodeck CH, Gill D, Rosenberg DA, et al. Testosterone Levels in Midtrimester Maternal and Fetal Plasma and Amniotic-Fluid. Prenatal Diag 1985;5:175-81
  • Dawood MY, Saxena BB. Testosterone and dihydrotestosterone in maternal and cord blood and in amniotic fluid. Am J Obstet Gynecol 1977;129:37-42
  • Makieva S, Saunders PT, Norman JE. Androgens in pregnancy: roles in parturition. Hum Reprod Update 2014;20:542-59
  • Yu CK, Papageorghiou AT, Bindra R, et al. Second-trimester sex hormone-binding globulin and subsequent development of pre-eclampsia. J Matern-Fetal Neo M 2004;16:158-62
  • Crespi EJ, Steckler TL, Mohankumar PS, et al. Prenatal exposure to excess testosterone modifies the developmental trajectory of the insulin-like growth factor system in female sheep. J Physiol 2006;572:119-30
  • Sathishkumar K, Elkins R, Chinnathambi V, et al. Prenatal testosterone-induced fetal growth restriction is associated with down-regulation of rat placental amino acid transport. Reprod Biol Endocrinol 2011;9:110
  • McCarthy A, Hughes R, Tilling K, et al. Birth weight; postnatal, infant, and childhood growth; and obesity in young adulthood: evidence from the Barry Caerphilly Growth Study. Am J Clin Nutr 2007;86:907-13
  • Voegtline KM, Costigan KA, Kivlighan KT, et al. Sex-specific associations of maternal prenatal testosterone levels with birth weight and weight gain in infancy. J Dev Orig Hlth Dis 2013;4:280-4
  • Carlsen SM, Jacobsen G, Vanky E. Mid-pregnancy androgen levels are negatively associated with breastfeeding. Acta Obstet Gyn Scan 2010;89:87-94
  • Dokras A, Spaczynski RZ, Behrman HP, et al. Testosterone levels in pregnant women correlate with the insulin response during the glucose tolerance test. Fertil Steril 2003;79:492-7
  • Troisi R, Innes KE, Roberts JM, et al. Preeclampsia and maternal breast cancer risk by offspring gender: do elevated androgen concentrations play a role? Brit J Cancer 2007;97:688-90
  • Uzelac PS, Li X, Lin J, et al. Dysregulation of Leptin and Testosterone Production and Their Receptor Expression in the Human Placenta with Gestational Diabetes Mellitus. Placenta 2010;31:581-8
  • Maliqueo M, Lara HE, Sanchez F, et al. Placental steroidogenesis in pregnant women with polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 2013;166:151-5
  • Sir-Petermann T, Maliqueo M, Angel B, et al. Maternal serum androgens in pregnant women with polycystic ovarian syndrome: possible implications in prenatal androgenization. Hum Reprod 2002;17:2573-9
  • Cannon B, Nedergaard J. Brown adipose tissue: Function and physiological significance. Physiol Rev 2004;84:277-359
  • Giralt M, Martin I, Iglesias R, et al. Ontogeny and perinatal modulation of gene expression in rat brown adipose tissue. Unaltered iodothyronine 5’-deiodinase activity is necessary for the response to environmental temperature at birth. Eur J Biochem 1990;193:297-302
  • Symonds ME, Lomax MA. Maternal and environmental influences on thermoregulation in the neonate. Proc Nutr Soc 1992;51:165-72
  • Clarke L, Heasman L, Firth K, et al. Influence of route of delivery and ambient temperature on thermoregulation in newborn lambs. Am J Physiol 1997;272:R1931-9
  • Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell 2007;131:242-56
  • Sondergaard E, Gormsen LC, Christensen MH, et al. Chronic adrenergic stimulation induces brown adipose tissue differentiation in visceral adipose tissue. Diabet Med 2015;32:e4-8
  • Girousse A, Langin D. Adipocyte lipases and lipid droplet-associated proteins: insight from transgenic mouse models. Int J Obes (Lond) 2012;36:581-94
  • Fruhbeck G, Mendez-Gimenez L, Fernandez-Formoso JA, et al. Regulation of adipocyte lipolysis. Nutr Res Rev 2014;27:63-93
  • Galic S, Oakhill JS, Steinberg GR. Adipose tissue as an endocrine organ. Mol Cell Endocrinol 2010;316:129-39
  • Hocking S, Samocha-Bonet D, Milner KL, et al. Adiposity and insulin resistance in humans: the role of the different tissue and cellular lipid depots. Endocr Rev 2013;34:463-500
  • Poissonnet CM, Burdi AR, Bookstein FL. Growth and development of human adipose tissue during early gestation. Early Hum Dev 1983;8:1-11
  • Poissonnet CM, Burdi AR, Garn SM. The chronology of adipose tissue appearance and distribution in the human fetus. Early Hum Dev 1984;10:1-11
  • Enzi G, Zanardo V, Caretta F, et al. Intrauterine growth and adipose tissue development. Am J Clin Nutr 1981;34:1785-90
  • Mostyn A, Bispham J, Pearce S, et al. Differential effects of leptin on thermoregulation and uncoupling protein abundance in the neonatal lamb. Faseb J 2002;16:1438-40
  • Smith SR, Lovejoy JC, Greenway F, et al. Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism 2001;50:425-35
  • Mayes JS, Watson GH. Direct effects of sex steroid hormones on adipose tissues and obesity. Obes Rev 2004;5:197-216
  • Rodriguez-Cuenca S, Monjo M, Proenza AM, et al. Depot differences in steroid receptor expression in adipose tissue: possible role of the local steroid milieu. Am J Physiol Endocrinol Metab 2005;288:E200-7
  • Joyner J, Hutley L, Cameron D. Intrinsic regional differences in androgen receptors and dihydrotestosterone metabolism in human preadipocytes. Horm Metab Res 2002;34:223-8
  • Bolt HM, Gobel P. Formation of estrogens from androgens by human subcutaneous adipose tissue in vitro. Horm Metab Res 1972;4:312-13
  • Escobar-Morreale HF, Alvarez-Blasco F, Botella-Carretero JI, et al. The striking similarities in the metabolic associations of female androgen excess and male androgen deficiency. Hum Reprod 2014;29:2083-91
  • Montes-Nieto R, Insenser M, Martinez-Garcia MA, et al. A nontargeted proteomic study of the influence of androgen excess on human visceral and subcutaneous adipose tissue proteomes. J Clin Endocr Metab 2013;98:E576-85
  • Varlamov O, White AE, Carroll JM, et al. Androgen effects on adipose tissue architecture and function in nonhuman primates. Endocrinology 2012;153:3100-10
  • van Houten EL, Kramer P, McLuskey A, et al. Reproductive and metabolic phenotype of a mouse model of PCOS. Endocrinology 2012;153:2861-9
  • Manneras L, Cajander S, Holmang A, et al. A new rat model exhibiting both ovarian and metabolic characteristics of polycystic ovary syndrome. Endocrinology 2007;148:3781-91
  • Blouin K, Nadeau M, Perreault M, et al. Effects of androgens on adipocyte differentiation and adipose tissue explant metabolism in men and women. Clin Endocrinol 2010;72:176-88
  • Chazenbalk G, Singh P, Irge D, et al. Androgens inhibit adipogenesis during human adipose stem cell commitment to preadipocyte formation. Steroids 2013;78:920-6
  • Gupta V, Bhasin S, Guo W, et al. Effects of dihydrotestosterone on differentiation and proliferation of human mesenchymal stem cells and preadipocytes. Mol Cell Endocrinol 2008;296:32-40
  • Barbosa-Desongles A, Hernandez C, Simo R, et al. Testosterone induces cell proliferation and cell cycle gene overexpression in human visceral preadipocytes. Am J Physiol Cell Physiol 2013;305:C355-9
  • Dicker A, Ryden M, Naslund E, et al. Effect of testosterone on lipolysis in human pre-adipocytes from different fat depots. Diabetologia 2004;47:420-8
  • Elbers JM, de Jong S, Teerlink T, et al. Changes in fat cell size and in vitro lipolytic activity of abdominal and gluteal adipocytes after a one-year cross-sex hormone administration in transsexuals. Metabolism 1999;48:1371-7
  • Corbould A. Chronic testosterone treatment induces selective insulin resistance in subcutaneous adipocytes of women. J Endocrinol 2007;192:585-94
  • Xu XF, De Pergola G, Bjorntorp P. Testosterone increases lipolysis and the number of beta-adrenoceptors in male rat adipocytes. Endocrinology 1991;128:379-82
  • Karbowska J, Kochan Z. Fat-reducing effects of dehydroepiandrosterone involve upregulation of ATGL and HSL expression, and stimulation of lipolysis in adipose tissue. Steroids 2012;77:1359-65
  • Nishizawa H, Shimomura H, Kishida K, et al. Androgens decrease plasma adiponectin, an insulin-sensitizing adipocyte-derived protein. Diabetes 2002;51:2734-41
  • Xu AM, Chan KW, Hoo RLC, et al. Testosterone selectively reduces the high molecular weight form of adiponectin by inhibiting its secretion from adipocytes. J Biol Chem 2005;280:18073-80
  • Luukkaa V, Pesonen U, Huhtaniemi I, et al. Inverse correlation between serum testosterone and leptin in men. J Clin Endocr Metab 1998;83:3243-6
  • Pineiro V, Casabiell X, Peino R, et al. Dihydrotestosterone, stanozolol, androstenedione and dehydroepiandrosterone sulphate inhibit leptin secretion in female but not in male samples of omental adipose tissue in vitro: lack of effect of testosterone. J Endocrinol 1999;160:425-32
  • Sato T, Matsumoto T, Yamada T, et al. Late onset of obesity in male androgen receptor-deficient (AR KO) mice. Biochem Biophys Res Commun 2003;300:167-71
  • Fan W, Yanase T, Nomura M, et al. Androgen receptor null male mice develop late-onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion. Diabetes 2005;54:1000-8
  • Lin HY, Xu Q, Yeh S, et al. Insulin and leptin resistance with hyperleptinemia in mice lacking androgen receptor. Diabetes 2005;54:1717-25
  • Rana K, Fam BC, Clarke MV, et al. Increased adiposity in DNA binding-dependent androgen receptor knockout male mice associated with decreased voluntary activity and not insulin resistance. Am J Physiol Endocrinol Metab 2011;301:E767-78
  • Rodriguez-Cuenca S, Pujol E, Justo R, et al. Sex-dependent thermogenesis, differences in mitochondrial morphology and function, and adrenergic response in brown adipose tissue. J Biol Chem 2002;277:42958-63
  • Rodriguez-Cuenca S, Monjo M, Frontera M, et al. Sex steroid receptor expression profile in brown adipose tissue. Effects of hormonal status. Cell Physiol Biochem 2007;20:877-86
  • Rodriguez AM, Monjo M, Roca P, et al. Opposite actions of testosterone and progesterone on UCP1 mRNA expression in cultured brown adipocytes. Cell Mol Life Sci 2002;59:1714-23
  • Rodriguez-Cuenca S, Monjo M, Gianotti M, et al. Expression of mitochondrial biogenesis-signaling factors in brown adipocytes is influenced specifically by 17 beta-estradiol, testosterone, and progesterone. Am J Physiol Endocrinol Metab 2007;292:E340-6
  • Kloting N, Bluher M. Adipocyte dysfunction, inflammation and metabolic syndrome. Rev Endocr Metab Dis 2014;15:277-87
  • Rudich A, Kanety H, Bashan N. Adipose stress-sensing kinases: linking obesity to malfunction. Trends Endocrinol Metab 2007;18:291-9
  • Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006;17:4-12
  • Kraegen EW, Cooney GJ. Free fatty acids and skeletal muscle insulin resistance. Curr Opin Lipidol 2008;19:235-41
  • Weyer C, Foley JE, Bogardus C, et al. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts Type II diabetes independent of insulin resistance. Diabetologia 2000;43:1498-506
  • Spalding KL, Arner E, Westermark PO, et al. Dynamics of fat cell turnover in humans. Nature 2008;453:783-7
  • Despres JP, Moorjani S, Lupien PJ, et al. Regional distribution of body fat, plasma lipoproteins, and cardiovascular disease. Arteriosclerosis 1990;10:497-511
  • Ross R, Aru J, Freeman J, et al. Abdominal adiposity and insulin resistance in obese men. Am J Physiol Endocrinol Metab 2002;282:E657-63
  • Ross R, Freeman J, Hudson R, et al. Abdominal obesity, muscle composition, and insulin resistance in premenopausal women. J Clin Endocr Metab 2002;87:5044-51
  • Arsenault BJ, Lemieux I, Despres JP, et al. The hypertriglyceridemic-waist phenotype and the risk of coronary artery disease: results from the EPIC-Norfolk prospective population study. CMAJ 2010;182:1427-32
  • Despres JP. Abdominal obesity as important component of insulin-resistance syndrome. Nutrition 1993;9:452-9
  • Kelley DE, Thaete FL, Troost F, et al. Subdivisions of subcutaneous abdominal adipose tissue and insulin resistance. Am J Physiol Endocrinol Metab 2000;278:E941-8
  • Terry RB, Stefanick ML, Haskell WL, et al. Contributions of regional adipose tissue depots to plasma lipoprotein concentrations in overweight men and women: possible protective effects of thigh fat. Metabolism 1991;40:733-40
  • Buemann B, Astrup A, Pedersen O, et al. Possible role of adiponectin and insulin sensitivity in mediating the favorable effects of lower body fat mass on blood lipids. J Clin Endocr Metab 2006;91:1698-704
  • Snijder MB, Visser M, Dekker JM, et al. Low subcutaneous thigh fat is a risk factor for unfavourable glucose and lipid levels, independently of high abdominal fat. The Health ABC Study. Diabetologia 2005;48:301-8
  • Boettcher M, Machann J, Stefan N, et al. Intermuscular Adipose Tissue (IMAT): Association With Other Adipose Tissue Compartments and Insulin Sensitivity. J Magn Reson Imaging 2009;29:1340-5
  • Walker GE, Verti B, Marzullo P, et al. Deep subcutaneous adipose tissue: a distinct abdominal adipose depot. Obesity 2007;15:1933-43
  • Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome--an allostatic perspective. Biochim Biophys Acta 2010;1801:338-49
  • Tan CY, Vidal-Puig A. Adipose tissue expandability: the metabolic problems of obesity may arise from the inability to become more obese. Biochem Soc Trans 2008;36:935-40
  • Jensen MD. Gender differences in regional fatty-acid metabolism before and after meal ingestion. J Clin Invest 1995;96:2297-303
  • Meek SE, Nair KS, Jensen MD. Insulin regulation of regional free fatty acid metabolism. Diabetes 1999;48:10-14
  • Koutsari C, Jensen MD. Free fatty acid metabolism in human obesity. J Lipid Res 2006;47:1643-50
  • Park HT, Lee ES, Cheon YP, et al. The relationship between fat depot-specific preadipocyte differentiation and metabolic syndrome in obese women. Clin Endocrinol 2012;76:59-66
  • Lund TD, Munson DJ, Haldy ME, et al. Dihydrotestosterone may inhibit hypothalamo-pituitary-adrenal activity by acting through estrogen receptor in the male mouse. Neurosci Lett 2004;365:43-7
  • Lund TD, Hinds LR, Handa RJ. The androgen 5 alpha-dihydrotestosterone and its metabolite 5 alpha-androstan-3 beta,17 beta-diol inhibit the hypothalamo pituitary-adrenal response to stress by acting through estrogen receptor beta-expressing neurons in the hypothalamus. J Neurosci 2006;26:1448-56
  • Steckelbroeck S, Jin Y, Gopishetty S, et al. Human cytosolic 3alpha-hydroxysteroid dehydrogenases of the aldo-keto reductase superfamily display significant 3beta-hydroxysteroid dehydrogenase activity: implications for steroid hormone metabolism and action. J Biol Chem 2004;279:10784-95
  • Stanbrough M, Bubley GJ, Ross K, et al. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res 2006;66:2815-25
  • Roland AV, Nunemaker CS, Keller SR, et al. Prenatal androgen exposure programs metabolic dysfunction in female mice. J Endocrinol 2010;207:213-23
  • Yan X, Dai X, Wang J, et al. Prenatal androgen excess programs metabolic derangements in pubertal female rats. J Endocrinol 2013;217:119-29
  • Sun M, Maliqueo M, Benrick A, et al. Maternal androgen excess reduces placental and fetal weights, increases placental steroidogenesis, and leads to long-term health effects in their female offspring. Am J Physiol Endocrinol Metab 2012;303:E1373-85
  • Lazic M, Aird F, Levine JE, et al. Prenatal androgen treatment alters body composition and glucose homeostasis in male rats. J Endocrinol 2011;208:293-300
  • Alexanderson C, Eriksson E, Stener-Victorin E, et al. Postnatal testosterone exposure results in insulin resistance, enlarged mesenteric adipocytes, and an atherogenic lipid profile in adult female rats: comparisons with estradiol and dihydrotestosterone. Endocrinology 2007;148:5369-76
  • Nohara K, Waraich RS, Liu S, et al. Developmental androgen excess programs sympathetic tone and adipose tissue dysfunction and predisposes to a cardiometabolic syndrome in female mice. Am J Physiol Endocrinol Metab 2013;304:E1321-30
  • Hansen LR, Drackley JK, Berger LL, et al. Prenatal androgenization of lambs: II. Metabolism in adipose tissue and liver. J Anim Sci 1995;73:1701-12
  • Nada SE, Thompson RC, Padmanabhan V. Developmental programming: differential effects of prenatal testosterone excess on insulin target tissues. Endocrinology 2010;151:5165-73
  • Veiga-Lopez A, Moeller J, Patel D, et al. Developmental programming: impact of prenatal testosterone excess on insulin sensitivity, adiposity, and free fatty acid profile in postpubertal female sheep. Endocrinology 2013;154:1731-42
  • Padmanabhan V, Veiga-Lopez A, Abbott DH, et al. Developmental programming: impact of prenatal testosterone excess and postnatal weight gain on insulin sensitivity index and transfer of traits to offspring of overweight females. Endocrinology 2010;151:595-605
  • Recabarren SE, Padmanabhan V, Codner E, et al. Postnatal developmental consequences of altered insulin sensitivity in female sheep treated prenatally with testosterone. Am J Physiol Endocrinol Metab 2005;289:E801-6
  • Hogg K, Wood C, McNeilly AS, et al. The in utero programming effect of increased maternal androgens and a direct fetal intervention on liver and metabolic function in adult sheep. PLoS ONE 2011;6:e24877
  • Bruns CM, Baum ST, Colman RJ, et al. Prenatal androgen excess negatively impacts body fat distribution in a nonhuman primate model of polycystic ovary syndrome. Int J Obes (Lond) 2007;31:1579-85
  • Keller E, Chazenbalk GD, Aguilera P, et al. Impaired preadipocyte differentiation into adipocytes in subcutaneous abdominal adipose of PCOS-like female rhesus monkeys. Endocrinology 2014;155:2696-703
  • Xu N, Kwon S, Abbott DH, et al. Epigenetic mechanism underlying the development of polycystic ovary syndrome (PCOS)-like phenotypes in prenatally androgenized rhesus monkeys. PLoS ONE 2011;6:e27286
  • Ogilvie CM, Crouch NS, Rumsby G, et al. Congenital adrenal hyperplasia in adults: a review of medical, surgical and psychological issues. Clin Endocrinol 2006;64:2-11
  • Cornean RE, Hindmarsh PC, Brook CGD. Obesity in 21-hydroxylase deficient patients. Arch Dis Child 1998;78:261-3
  • Goodarzi MO, Dumesic DA, Chazenbalk G, et al. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat Rev Endocrinol 2011;7:219-31
  • Sir-Petermann T, Maliqueo M, Codner E, et al. Early metabolic derangements in daughters of women with polycystic ovary syndrome. J Clin Endocr Metab 2007;92:4637-42
  • Trottier A, Battista MC, Geller DH, et al. Adipose tissue insulin resistance in peripubertal girls with first-degree family history of polycystic ovary syndrome. Fertil Steril 2012;98:1627-34
  • Recabarren SE, Smith R, Rios R, et al. Metabolic profile in sons of women with polycystic ovary syndrome. J Clin Endocr Metab 2008;93:1820-6
  • Villa J, Pratley RE. Adipose tissue dysfunction in polycystic ovary syndrome. Curr Diab Rep 2011;11:179-84
  • Ek I, Arner P, Bergqvist A, et al. Impaired adipocyte lipolysis in nonobese women with the polycystic ovary syndrome: a possible link to insulin resistance? J Clin Endocr Metab 1997;82:1147-53
  • Faulds G, Ryden M, Ek I, et al. Mechanisms behind lipolytic catecholamine resistance of subcutaneous fat cells in the polycystic ovarian syndrome. J Clin Endocr Metab 2003;88:2269-73
  • Manneras-Holm L, Leonhardt H, Kullberg J, et al. Adipose Tissue Has Aberrant Morphology and Function in PCOS: Enlarged Adipocytes and Low Serum Adiponectin, But Not Circulating Sex Steroids, Are Strongly Associated with Insulin Resistance. J Clin Endocr Metab 2011;96:E304-11
  • Rosenbaum D, Haber RS, Dunaif A. Insulin resistance in polycystic ovary syndrome: decreased expression of GLUT-4 glucose transporters in adipocytes. Am J Physiol 1993;264:E197-202
  • de Zegher F, Lopez-Bermejo A, Ibanez L. Adipose tissue expandability and the early origins of PCOS. Trends Endocrinol Metab 2009;20:418-23

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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