Horm Metab Res 2012; 44(08): 598-606
DOI: 10.1055/s-0032-1311576
Review
© Georg Thieme Verlag KG Stuttgart · New York

Models and Mechanisms of Metabolic Regulation: Genes, Stress, and the HPA and HPG Axes

G. J. Boersma
1   Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
,
S. R. Salton
2   Department of Neuroscience, Mount Sinai School of Medicine, New York, USA
,
P. M. Spritzer
3   Departamento de Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
,
C. T. Steele
4   Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda MD, USA
,
D. L. Carbone
5   Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
› Author Affiliations
Further Information

Publication History

received 09 December 2011

accepted 15 March 2012

Publication Date:
27 April 2012 (online)

Abstract

A variety of models have been developed to better understand the mechanisms underlying individual variation in susceptibility to obesity. This review discusses several of these models and explores their role in understanding individual vulnerability to metabolic disease and the environmental factors around which metabolic perturbations occur. Recently, the focus of models has shifted towards heterogeneous populations, in which individuals characterized by a high vulnerability and individuals that are seemingly resistant can be identified. The use of these heterogeneous studies has lead to the identification of several novel biomarkers predicting obesity. This review therefore focuses on nontraditional factors, which are not directly implicated in metabolic regulation. First, the evidence from rodent knockout models for genetic factors involved in obesity is discussed. Second, the role of a stressful environment, particularly the early life environment is investigated along with a discussion of circadian disruption and metabolic disorders. Finally, the impact of sex-steroids, as exemplified by polycystic ovarian syndrome, is discussed. Overall, the data presented in our review demonstrate that in most cases interplay between genetic and environmental factors best predicts disease development. Our review shows that susceptibility to obesity may be explained by complex interactions between traditional homeostatic mechanisms, such as the hypothalamic peptide, and less studied mechanisms, like steroids and neurotrophic factors.

 
  • References

  • 1 World Health O . world health statistics http://www.who.int/whosis/whostat/EN_WHS10_Full.pdf
  • 2 Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia 1992; 35: 595-601
  • 3 Bartness TJ, Song CK, Demas GE. SCN efferents to peripheral tissues: implications for biological rhythms. J Biol Rhythms 2001; 16: 196-204
  • 4 Barsh G. From Agouti to Pomc–100 years of fat blonde mice. Nat Med 1999; 5: 984-985
  • 5 Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 1997; 88: 131-141
  • 6 Wortley KE, Anderson KD, Yasenchak J, Murphy A, Valenzuela D, Diano S, Yancopoulos GD, Wiegand SJ, Sleeman MW. Agouti-related protein-deficient mice display an age-related lean phenotype. Cell Metab 2005; 2: 421-427
  • 7 Flier JS. AgRP in energy balance: Will the real AgRP please stand up?. Cell Metab 2006; 3: 83-85
  • 8 Luo N, Marcelin G, Liu SM, Schwartz G, Chua Jr S. Neuropeptide Y and agouti-related peptide mediate complementary functions of hyperphagia and reduced energy expenditure in leptin receptor deficiency. Endocrinology 2011; 152: 883-889
  • 9 Cota D, Marsicano G, Tschop M, Grubler Y, Flachskamm C, Schubert M, Auer D, Yassouridis A, Thone-Reineke C, Ortmann S, Tomassoni F, Cervino C, Nisoli E, Linthorst AC, Pasquali R, Lutz B, Stalla GK, Pagotto U. The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J Clin Invest 2003; 112: 423-431
  • 10 Quarta C, Bellocchio L, Mancini G, Mazza R, Cervino C, Braulke LJ, Fekete C, Latorre R, Nanni C, Bucci M, Clemens LE, Heldmaier G, Watanabe M, Leste-Lassere T, Maitre M, Tedesco L, Fanelli F, Reuss S, Klaus S, Srivastava RK, Monory K, Valerio A, Grandis A, De Giorgio R, Pasquali R, Nisoli E, Cota D, Lutz B, Marsicano G, Pagotto U. CB(1) signaling in forebrain and sympathetic neurons is a key determinant of endocannabinoid actions on energy balance. Cell Metab 2010; 11: 273-285
  • 11 Carlin KM, Vale WW, Bale TL. Vital functions of corticotropin-releasing factor (CRF) pathways in maintenance and regulation of energy homeostasis. Proc Natl Acad Sci USA 2006; 103: 3462-3467
  • 12 Sakurai T, Mieda M. Connectomics of orexin-producing neurons: interface of systems of emotion, energy homeostasis and arousal. Trends Pharmacol Sci 2011; 32: 451-462
  • 13 Levin BE. Neurotrophism and energy homeostasis: perfect together. Am J Physiol Regul Integr Comp Physiol 2007; 293: R988-R991
  • 14 Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and locomotor activity in mice. Embo J 2000; 19: 1290-1300
  • 15 Rios M, Fan G, Fekete C, Kelly J, Bates B, Kuehn R, Lechan RM, Jaenisch R. Conditional deletion of brain-derived neurotrophic factor in the postnatal brain leads to obesity and hyperactivity. Mol Endocrinol 2001; 15: 1748-1757
  • 16 Unger TJ, Calderon GA, Bradley LC, Sena-Esteves M, Rios M. Selective deletion of Bdnf in the ventromedial and dorsomedial hypothalamus of adult mice results in hyperphagic behavior and obesity. J Neurosci 2007; 27: 14265-14274
  • 17 Wang C, Bomberg E, Billington C, Levine A, Kotz CM. Brain-derived neurotrophic factor in the hypothalamic paraventricular nucleus increases energy expenditure by elevating metabolic rate. Am J Physiol Regul Integr Comp Physiol 2007; 293: R992-R1002
  • 18 Holtmann B, Wiese S, Samsam M, Grohmann K, Pennica D, Martini R, Sendtner M. Triple knock-out of CNTF, LIF, and CT-1 defines cooperative and distinct roles of these neurotrophic factors for motoneuron maintenance and function. J Neurosci 2005; 25: 1778-1787
  • 19 Kokoeva MV, Yin H, Flier JS. Neurogenesis in the hypothalamus of adult mice: potential role in energy balance. Science 2005; 310: 679-683
  • 20 Domouzoglou EM, Maratos-Flier E. Fibroblast growth factor 21 is a metabolic regulator that plays a role in the adaptation to ketosis. Am J Clin Nutr 2011; 93: 901S-905S
  • 21 Potthoff MJ, Inagaki T, Satapati S, Ding X, He T, Goetz R, Mohammadi M, Finck BN, Mangelsdorf DJ, Kliewer SA, Burgess SC. FGF21 induces PGC-1alpha and regulates carbohydrate and fatty acid metabolism during the adaptive starvation response. Proc Natl Acad Sci USA 2009; 106: 10853-10858
  • 22 Song CK, Jackson RM, Harris RB, Richard D, Bartness TJ. Melanocortin-4 receptor mRNA is expressed in sympathetic nervous system outflow neurons to white adipose tissue. Am J Physiol Regul Integr Comp Physiol 2005; 289: R1467-R1476
  • 23 Voss-Andreae A, Murphy JG, Ellacott KL, Stuart RC, Nillni EA, Cone RD, Fan W. Role of the central melanocortin circuitry in adaptive thermogenesis of brown adipose tissue. Endocrinology 2007; 148: 1550-1560
  • 24 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
  • 25 Kishi T, Aschkenasi CJ, Lee CE, Mountjoy KG, Saper CB, Elmquist JK. Expression of melanocortin 4 receptor mRNA in the central nervous system of the rat. J Comp Neurol 2003; 457: 213-235
  • 26 Collins S, Cao W, Robidoux J. Learning new tricks from old dogs: beta-adrenergic receptors teach new lessons on firing up adipose tissue metabolism. Mol Endocrinol 2004; 18: 2123-2131
  • 27 Turtzo LC, Marx R, Lane MD. Cross-talk between sympathetic neurons and adipocytes in coculture. Proc Natl Acad Sci USA 2001; 98: 12385-12390
  • 28 Thomas SA, Palmiter RD. Thermoregulatory and metabolic phenotypes of mice lacking noradrenaline and adrenaline. Nature 1997; 387: 94-97
  • 29 Bachman ES, Dhillon H, Zhang CY, Cinti S, Bianco AC, Kobilka BK, Lowell BB. betaAR signaling required for diet-induced thermogenesis and obesity resistance. Science 2002; 297: 843-845
  • 30 Bartolomucci A, Possenti R, Mahata SK, Fischer-Colbrie R, Loh YP, Salton SR. The extended granin family: structure, function, and biomedical implications. Endocr Rev 2011; 32: 755-797
  • 31 Clarke IJ. Models of ‘obesity’ in large animals and birds. Front Horm Res 2008; 36: 107-117
  • 32 Patterson CM, Bouret SG, Dunn-Meynell AA, Levin BE. Three weeks of postweaning exercise in DIO rats produces prolonged increases in central leptin sensitivity and signaling. Am J Physiol Regul Integr Comp Physiol 2009; 296: R537-R548
  • 33 Buren J, Eriksson JW. Is insulin resistance caused by defects in insulin’s target cells or by a stressed mind?. Diabetes Metab Res Rev 2005; 21: 487
  • 34 Munir A, Newell-Price J. Management of diabetes mellitus in Cushing’s syndrome. Neuroendocrinology 2010; 92 (Suppl. 01) 82-85
  • 35 Chan O, Inouye K, Riddell MC, Vranic M, Matthews SG. Diabetes and the hypothalamo-pituitary-adrenal (HPA) axis. Minerva Endocrinol 2003; 28: 87-102
  • 36 Koolhaas JM, Korte SM, de Boer SF, van der Vegt BJ, Van Reenen CG, Hopster H, De JI, Ruis MA, Blokhuis HJ. Coping styles in animals: current status in behavior and stress-physiology. Neurosci Biobehav Rev 1999; 23: 925
  • 37 Boersma GJ, Benthem L, van DG, Scheurink AJ. Individual variation in the (patho)physiology of energy balance. Physiol Behav 2011; 103: 89-97
  • 38 Boersma G, Benthem L, van DG, Steimer TJ, Scheurink AJ. Pharmacological treatment of hyperinsulineamia in rats depends on coping style. Eur J Pharmacol 2011; 654: 122-127
  • 39 Dhabhar FS, McEwen BS, Spencer RL. Stress response, adrenal steroid receptor levels and corticosteroid-binding globulin levels–a comparison between Sprague-Dawley, Fischer 344 and Lewis rats. Brain Res 1993; 616: 89
  • 40 Fink RI, Huecksteadt T, Karaoghlanian Z. The effects of aging on glucose metabolism in adipocytes from Fischer rats. Endocrinology 1986; 118: 1139-1147
  • 41 Combettes M, Kargar C. Newly approved and promising antidiabetic agents. Therapie 2007; 62: 293
  • 42 Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ 1989; 298: 564-567
  • 43 Reinisch JM, Simon NG, Karow WG, Gandelman R. Prenatal exposure to prednisone in humans and animals retards intrauterine growth. Science 1978; 202: 436-438
  • 44 Mericq V, Medina P, Kakarieka E, Marquez L, Johnson MC, Iniguez G. Differences in expression and activity of 11beta-hydroxysteroid dehydrogenase type 1 and 2 in human placentas of term pregnancies according to birth weight and gender. Eur J Endocrinol 2009; 161: 419-425
  • 45 McTernan CL, Draper N, Nicholson H, Chalder SM, Driver P, Hewison M, Kilby MD, Stewart PM. Reduced placental 11beta-hydroxysteroid dehydrogenase type 2 mRNA levels in human pregnancies complicated by intrauterine growth restriction: an analysis of possible mechanisms. J Clin Endocrinol Metab 2001; 86: 4979-4983
  • 46 Stewart PM, Rogerson FM, Mason JI. Type 2 11 beta-hydroxysteroid dehydrogenase messenger ribonucleic acid and activity in human placenta and fetal membranes: its relationship to birth weight and putative role in fetal adrenal steroidogenesis. J Clin Endocrinol Metab 1995; 80: 885-890
  • 47 Struwe E, Berzl GM, Schild RL, Beckmann MW, Dorr HG, Rascher W, Dotsch J. Simultaneously reduced gene expression of cortisol-activating and cortisol-inactivating enzymes in placentas of small-for-gestational-age neonates. Am J Obstet Gynecol 2007; 197 (43) e41-e46
  • 48 Benediktsson R, Lindsay RS, Noble J, Seckl JR, Edwards CR. Glucocorticoid exposure in utero: new model for adult hypertension. Lancet 1993; 341: 339-341
  • 49 Lindsay RS, Lindsay RM, Waddell BJ, Seckl JR. Prenatal glucocorticoid exposure leads to offspring hyperglycaemia in the rat: studies with the 11 beta-hydroxysteroid dehydrogenase inhibitor carbenoxolone. Diabetologia 1996; 39: 1299-1305
  • 50 Copper RL, Goldenberg RL, Das A, Elder N, Swain M, Norman G, Ramsey R, Cotroneo P, Collins BA, Johnson F, Jones P, Meier AM. The preterm prediction study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks’ gestation. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Am J Obstet Gynecol 1996; 175: 1286-1292
  • 51 Szuran T, Zimmerman E, Pliska V, Pfister HP, Welzl H. Prenatal stress effects on exploratory activity and stress-induced analgesia in rats. Dev Psychobiol 1991; 24: 361-372
  • 52 Weinstock M, Poltyrev T, Schorer-Apelbaum D, Men D, McCarty R. Effect of prenatal stress on plasma corticosterone and catecholamines in response to footshock in rats. Physiol Behav 1998; 64: 439-444
  • 53 Maccari S, Morley-Fletcher S. Effects of prenatal restraint stress on the hypothalamus-pituitary-adrenal axis and related behavioural and neurobiological alterations. Psychoneuroendocrinology 2007; 32 (Suppl. 01) S10-S15
  • 54 Vanbesien-Mailliot CC, Wolowczuk I, Mairesse J, Viltart O, Delacre M, Khalife J, Chartier-Harlin MC, Maccari S. Prenatal stress has pro-inflammatory consequences on the immune system in adult rats. Psychoneuroendocrinology 2007; 32: 114-124
  • 55 Desai M, Gayle D, Babu J, Ross MG. Programmed obesity in intrauterine growth-restricted newborns: modulation by newborn nutrition. Am J Physiol Regul Integr Comp Physiol 2005; 288: R91
  • 56 Patel MS, Srinivasan M, Laychock SG. Metabolic programming: Role of nutrition in the immediate postnatal life. J Inherit Metab Dis 2009; 32: 218
  • 57 Tamashiro KL, Moran TH. Perinatal environment and its influences on metabolic programming of offspring. Physiol Behav 2010; 100: 560-566
  • 58 Norberg H, Stalnacke J, Heijtz RD, Smedler AC, Nyman M, Forssberg H, Norman M. Antenatal corticosteroids for preterm birth: dose-dependent reduction in birthweight, length and head circumference. Acta Paediatr 2011; 100: 364-369
  • 59 Watterberg KL. Policy statement–postnatal corticosteroids to prevent or treat bronchopulmonary dysplasia. Pediatrics 2010; 126: 800-808
  • 60 Cleasby ME, Kelly PA, Walker BR, Seckl JR. Programming of rat muscle and fat metabolism by in utero overexposure to glucocorticoids. Endocrinology 2003; 144: 999-1007
  • 61 Coe CL, Lubach GR. Developmental consequences of antenatal dexamethasone treatment in nonhuman primates. Neurosci Biobehav Rev 2005; 29: 227-235
  • 62 Drake AJ, Raubenheimer PJ, Kerrigan D, McInnes KJ, Seckl JR, Walker BR. Prenatal dexamethasone programs expression of genes in liver and adipose tissue and increased hepatic lipid accumulation but not obesity on a high-fat diet. Endocrinology 2010; 151: 1581-1587
  • 63 McKendry AA, Palliser HK, Yates DM, Walker DW, Hirst JJ. The effect of betamethasone treatment on neuroactive steroid synthesis in a foetal Guinea pig model of growth restriction. J Neuroendocrinol 2010; 22: 166-174
  • 64 Hossain A, Hajman K, Charitidi K, Erhardt S, Zimmermann U, Knipper M, Canlon B. Prenatal dexamethasone impairs behavior and the activation of the BDNF exon IV promoter in the paraventricular nucleus in adult offspring. Endocrinology 2008; 149: 6356-6365
  • 65 Shoener JA, Baig R, Page KC. Prenatal exposure to dexamethasone alters hippocampal drive on hypothalamic-pituitary-adrenal axis activity in adult male rats. Am J Physiol Regul Integr Comp Physiol 2006; 290: R1366-R1373
  • 66 O’Regan D, Kenyon CJ, Seckl JR, Holmes MC. Prenatal dexamethasone ‘programmes’ hypotension, but stress-induced hypertension in adult offspring. J Endocrinol 2008; 196: 343-352
  • 67 Bass TJ. Circadian Integration of Metabolism and Energetics. Science 2010; 330: 1349-1344
  • 68 Scheer FA, Hilton MF, Mantzoros CS, Shea SA. Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA 2009; 106: 4453-4458
  • 69 van Mark ASM, Kessel R, Brinkmann E. Shift work and pathological conditions. J Occup Med Toxicol 2006; 1
  • 70 Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment of the circadian clock in the liver by feeding. Science 2001; 291: 490-493
  • 71 Mustonen AM, Nieminen P, Hyvarinen H. Effects of continuous light and melatonin treatment on energy metabolism of the rat. J Endocrinol Invest 2001; 25: 716-723
  • 72 Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A, Losee-Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J. Obesity and metabolic syndrome in circadian Clock mutant mice. Science 2005; 308: 1043-1045
  • 73 Duez H, Staels B. Rev-erb-alpha: an integrator of circadian rhythms and metabolism. J Appl Physiol 2009; 107: 1972-1980
  • 74 Le Martelot G, Claudel T, Gatfield D, Schaad O, Kornmann B, Sasso GL, Moschetta A, Schibler U. REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol 2009; 7: e1000181
  • 75 Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS). Hum Reprod 2004; 19: 41-47
  • 76 Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocrinol Metab 2004; 89: 2745-2749
  • 77 Azziz R, Sanchez LA, Knochenhauer ES, Moran C, Lazenby J, Stephens KC, Taylor K, Boots LR. Androgen excess in women: experience with over 1 000 consecutive patients. J Clin Endocrinol Metab 2004; 89: 453-462
  • 78 Toscani M, Migliavacca R, Sisson de Castro JA, Spritzer PM. Estimation of truncal adiposity using waist circumference or the sum of trunk skinfolds: a pilot study for insulin resistance screening in hirsute patients with or without polycystic ovary syndrome. Metabolism 2007; 56: 992-997
  • 79 Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and predictors of risk for type 2 diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospective, controlled study in 254 affected women. J Clin Endocrinol Metab 1999; 84: 165-169
  • 80 Spritzer PM, Wiltgen D. Prevalence of metabolic syndrome in patients of south of Brazil with polycystic ovary syndrome (PCOS). Arq Bras Endocrinol Metabol 2007; 51: 146-147
  • 81 Bjorntorp P. Body fat distribution, insulin resistance, and metabolic diseases. Nutrition 1997; 13: 795-803
  • 82 Mayes JS, Watson GH. Direct effects of sex steroid hormones on adipose tissues and obesity. Obes Rev 2004; 5: 197-216
  • 83 Wild RA, Carmina E, Diamanti-Kandarakis E, Dokras A, Escobar-Morreale HF, Futterweit W, Lobo R, Norman RJ, Talbott E, Dumesic DA. Assessment of cardiovascular risk and prevention of cardiovascular disease in women with the polycystic ovary syndrome: a consensus statement by the Androgen Excess and Polycystic Ovary Syndrome (AE-PCOS) Society. J Clin Endocrinol Metab 2010; 95: 2038-2049
  • 84 Maturana MA, Rubira MC, Consolim-Colombo F, Irigoyen MC, Spritzer PM. Androgenicity and venous endothelial function in post-menopausal women. J Endocrinol Invest 2010; 33: 239-243
  • 85 Maturana MA, Spritzer PM. Association between hyperinsulinemia and endogenous androgen levels in peri- and postmenopausal women. Metabolism 2001; 51: 238-243
  • 86 Shaw LJ, Bairey Merz CN, Azziz R, Stanczyk FZ, Sopko G, Braunstein GD, Kelsey SF, Kip KE, Cooper-Dehoff RM, Johnson BD, Vaccarino V, Reis SE, Bittner V, Hodgson TK, Rogers W, Pepine CJ. Postmenopausal women with a history of irregular menses and elevated androgen measurements at high risk for worsening cardiovascular event-free survival: results from the National Institutes of Health – National Heart, Lung, and Blood Institute sponsored Women’s Ischemia Syndrome Evaluation. J Clin Endocrinol Metab 2008; 93: 1276-1284
  • 87 Legro RS, Driscoll D, Strauss 3rd JF, Fox J, Dunaif A. Evidence for a genetic basis for hyperandrogenemia in polycystic ovary syndrome. Proc Natl Acad Sci USA 1998; 95: 14956-14960
  • 88 Kahsar-Miller MD, Nixon C, Boots LR, Go RC, Azziz R. Prevalence of polycystic ovary syndrome (PCOS) in first-degree relatives of patients with PCOS. Fertil Steril 2001; 75: 53-58
  • 89 Yildiz BO, Yarali H, Oguz H, Bayraktar M. Glucose intolerance, insulin resistance, and hyperandrogenemia in first degree relatives of women with polycystic ovary syndrome. J Clin Endocrinol Metab 2003; 88: 2031-2036
  • 90 Pasquali R, Stener-Victorin E, Yildiz BO, Duleba AJ, Hoeger K, Mason H, Homburg R, Hickey T, Franks S, Tapanainen JS, Balen A, Abbott DH, Diamanti-Kandarakis E, Legro RS. PCOS Forum: research in polycystic ovary syndrome today and tomorrow. Clin Endocrinol (Oxf) 2011; 74: 424-433
  • 91 Xita N, Tsatsoulis A. Review: fetal programming of polycystic ovary syndrome by androgen excess: evidence from experimental, clinical, and genetic association studies. J Clin Endocrinol Metab 2006; 91: 1660-1666
  • 92 Abbott DH, Barnett DK, Bruns CM, Dumesic DA. Androgen excess fetal programming of female reproduction: a developmental aetiology for polycystic ovary syndrome?. Hum Reprod Update 2005; 11: 357-374
  • 93 Barnes RB, Rosenfield RL, Ehrmann DA, Cara JF, Cuttler L, Levitsky LL, Rosenthal IM. Ovarian hyperandrogynism as a result of congenital adrenal virilizing disorders: evidence for perinatal masculinization of neuroendocrine function in women. J Clin Endocrinol Metab 1994; 79: 1328-1333
  • 94 Hague WM, Adams J, Rodda C, Brook CG, de Bruyn R, Grant DB, Jacobs HS. The prevalence of polycystic ovaries in patients with congenital adrenal hyperplasia and their close relatives. Clin Endocrinol (Oxf) 1990; 33: 501-510
  • 95 Xita N, Tsatsoulis A. Fetal origins of the metabolic syndrome. Ann N Y Acad Sci 2010; 1205: 148-155
  • 96 Blouin K, Nadeau M, Mailloux J, Daris M, Lebel S, Luu-The V, Tchernof A. Pathways of adipose tissue androgen metabolism in women: depot differences and modulation by adipogenesis. Am J Physiol Endocrinol Metab 2009; 296: E244-E255
  • 97 Lecke SB, Mattei F, Morsch DM, Spritzer PM. Abdominal subcutaneous fat gene expression and circulating levels of leptin and adiponectin in polycystic ovary syndrome. Fertil Steril 2011; 95: 2044-2049
  • 98 Lecke SB, Morsch DM, Spritzer PM. CYP19 gene expression in subcutaneous adipose tissue is associated with blood pressure in women with polycystic ovary syndrome. Steroids 2011; 76: 1383-1388
  • 99 Moran LJ, Hutchison SK, Norman RJ, Teede HJ. Lifestyle changes in women with polycystic ovary syndrome. Cochrane Database Syst Rev 2011; 16 CD007506