Horm Metab Res 2016; 48(03): 178-184
DOI: 10.1055/s-0035-1555902
Endocrine Care
© Georg Thieme Verlag KG Stuttgart · New York

Dysregulation of Hypothalamo-Pituitary-Adrenocortical Axis in Overweight Female Diabetic Subjects is Associated with Downregulation of Corticosteroid Receptors and 11β-HSD1 in the Brain

S. Li*
,
Y. Liao*
,
L. Wang
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
R. Huang
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
J. Yue
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
H. Xu
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
H. Zhou
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
Z. Lou
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
Y. Hu
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
,
W. Liu
1   Department of Endocrinology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University. Dongfang Road, Shanghai, China
› Author Affiliations
Further Information

Publication History

received 18 January 2015

accepted 25 June 2015

Publication Date:
24 July 2015 (online)

Abstract

The objective of this work was to assess hypothalamo-pituitary-adrenocortical (HPA) axis dysregulation in overweight diabetic women and investigate the possible mechanism using overweight diabetic rats. Twenty-two overweight diabetic women were recruited alongside 34 lean and 23 overweight healthy women serving as controls. Dexamethasone suppression test (0.25 mg DST) and low dose adrenocorticotropic hormone (ACTH) stimulation assay were used to evaluate the HPA axis activity. Then, high fat diet (HF) and STZ-induced diabetic rats were utilized to investigate the possible mechanism. After measurement of corticosterone circadian patterns and dexamethasone suppression levels, mRNA amounts of mineralocorticoid receptors (MR), glucocorticoid receptors (GR), and 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) were determined by real time PCR at hippocampus, hypothalamus, and pituitary levels. Overweight diabetic women showed impaired HPA axis with negative feedback efficacy (suppression ratio F-DEX%: 0.52±0.06% vs. 0.49±0.06% vs. 0.14±0.08%), as well as increased adrenal cortisol secretion response to low dose ACTH stimulation. Interestingly, F-DEX% was negatively correlated with BMI (r=− 0.323, p=0.003), waist circumference (r=− 0.319, p=0.004), and HbA1c (r=− 0.334, p=0.002). Stepwise linear regression analysis showed F-DEX% was significantly related to HbA1c level (β=− 0.328, p=0.007) after adjusting for other covariates (age, BMI, waist circumference, SBP, TC, TG, and HOMA-IR). Furthermore, 11β-HSD1, MR, and GR mRNA expression levels were reduced at pituitary level while GR expression was downregulated at hippocampus level in HF and HF+STZ rats. In conclusion, hyperactive HPA axis in overweight diabetic subjects may be associated with downregulation of 11β-HSD1, MR, and GR in the brain.

* These authors contributed equally to this work.


 
  • References

  • 1 Cameron OG, Kronfol Z, Greden JF, Carroll BJ. Hypothalamic-pituitary-adrenocortical activity in patients with diabetes mellitus. Arch Gen Psychiatr 1984; 41: 1090-1095
  • 2 Roy MS, Roy A, Gallucci WT, Collier B, Young K, Kamilaris TC, Chrousos GP. The ovine corticotropin-releasing hormone-stimulation test in type I diabetic patients and controls: suggestion of mild chronic hypercortisolism. Metab: Clin Exp 1993; 42: 696-700
  • 3 Roy M, Collier B, Roy A. Hypothalamic-pituitary-adrenal axis dysregulation among diabetic outpatients. Psychiatr Res 1990; 31: 31-37
  • 4 Asfeldt VH. Hypophyseo-adrenocortical function in diabetes mellitus. Acta Med Scand 1972; 191: 349-354
  • 5 Serio M, Tarquini B, Contini P, Bucalossi A, Toccafondi R. Plasma cortisol response to insulin and circadian rhythm in diabetic subjects. Diabetes 1968; 17: 124-126
  • 6 Bhatia RP, Adarsh Singh RH. Cortisol in diabetic retinopathy. Ann Ophthalmol 1983; 15: 128-130
  • 7 Tsigos C, Young RJ, White A. Diabetic neuropathy is associated with increased activity of the hypothalamic-pituitary-adrenal axis. J Clin Endocrinol Metab 1993; 76: 554-558
  • 8 Kopelman PG. Is the hypothalamic-pituitary-adrenal axis really hyperactivated in visceral obesity?. J Endocrinol Invest 1999; 22: 76-79
  • 9 Abraham SB, Rubino D, Sinaii N, Ramsey S, Nieman LK. Cortisol, obesity, and the metabolic syndrome: a cross-sectional study of obese subjects and review of the literature. Obesity 2013; 21: E105-E117
  • 10 Weaver JU, Kopelman PG, McLoughlin L, Forsling ML, Grossman A. Hyperactivity of the hypothalamo-pituitary-adrenal axis in obesity: a study of ACTH, AVP, beta-lipotrophin and cortisol responses to insulin-induced hypoglycaemia. Clin Endocrinol 1993; 39: 345-350
  • 11 Kjolhede EA, Gustafsson PE, Gustafsson PA, Nelson N. Overweight and obese children have lower cortisol levels than normal weight children. Acta Paediatr 2014; 103: 295-299
  • 12 Champaneri S, Xu X, Carnethon MR, Bertoni AG, Seeman T, DeSantis AS, Diez Roux A, Shrager S, Golden SH. Diurnal salivary cortisol is associated with body mass index and waist circumference: the Multiethnic Study of Atherosclerosis. Obesity 2013; 21: E56-E63
  • 13 Hudson JI, Hudson MS, Rothschild AJ, Vignati L, Schatzberg AF, Melby JC. Abnormal results of dexamethasone suppression tests in nondepressed patients with diabetes mellitus. Arch Gen Psychiatr 1984; 41: 1086-1089
  • 14 Ljung T, Andersson B, Bengtsson BA, Bjorntorp P, Marin P. Inhibition of cortisol secretion by dexamethasone in relation to body fat distribution: a dose-response study. Obesity Res 1996; 4: 277-282
  • 15 De Kloet ER, Vreugdenhil E, Oitzl MS, Joels M. Brain corticosteroid receptor balance in health and disease. Endocr Rev 1998; 19: 269-301
  • 16 Atanasov AG, Nashev LG, Schweizer RA, Frick C, Odermatt A. Hexose-6-phosphate dehydrogenase determines the reaction direction of 11beta-hydroxysteroid dehydrogenase type 1 as an oxoreductase. FEBS Lett 2004; 571: 129-133
  • 17 Atanasov AG, Odermatt A. Readjusting the glucocorticoid balance: an opportunity for modulators of 11beta-hydroxysteroid dehydrogenase type 1 activity?. Endocr Metab Immune Disorders Drug Targets 2007; 7: 125-140
  • 18 McEwen BS, Lambdin LT, Rainbow TC, De Nicola AF. Aldosterone effects on salt appetite in adrenalectomized rats. Neuroendocrinology 1986; 43: 38-43
  • 19 Li S, Liu W, Wang L, Huang R, Chen Q, Wu Y, Cai Y. Effects of menopause on hepatic 11beta-hydroxysteroid dehydrogenase type 1 activity and adrenal sensitivity to adrenocorticotropin in healthy non-obese women. Gynecol Endocrinol 2011; 27: 794-799
  • 20 Chan O, Inouye K, Vranic M, Matthews SG. Hyperactivation of the hypothalamo-pituitary-adrenocortical axis in streptozotocin-diabetes is associated with reduced stress responsiveness and decreased pituitary and adrenal sensitivity. Endocrinology 2002; 143: 1761-1768
  • 21 Pascoe WS, Smythe GA, Storlien LH. Enhanced responses to stress induced by fat-feeding in rats: relationship between hypothalamic noradrenaline and blood glucose. Brain Res 1991; 550: 192-196
  • 22 Chiodini I, Adda G, Scillitani A, Coletti F, Morelli V, Di Lembo S, Epaminonda P, Masserini B, Beck-Peccoz P, Orsi E, Ambrosi B, Arosio M. Cortisol secretion in patients with type 2 diabetes: relationship with chronic complications. Diabetes Care 2007; 30: 83-88
  • 23 Cooper ME. Pathogenesis, prevention, and treatment of diabetic nephropathy. Lancet 1998; 352: 213-219
  • 24 Prpic-Krizevac I, Canecki-Varzic S, Bilic-Curcic I. Hyperactivity of the hypothalamic-pituitary-adrenal axis in patients with type 2 diabetes and relations with insulin resistance and chronic complications. Wiener Klin Wochenschr 2012; 124: 403-411
  • 25 Ueshiba H, Shimizu Y, Hiroi N, Yakushiji F, Shimojo M, Tsuboi K, Miyachi Y. Decreased steroidogenic enzyme 17,20-lyase and increased 17-hydroxylase activities in type 2 diabetes mellitus. Eur J Endocrinol 2002; 146: 375-380
  • 26 Elahi-Moghaddam Z, Behnam-Rassouli M, Mahdavi-Shahri N, Hajinejad-Boshroue R, Khajouee E. Comparative study on the effects of type 1 and type 2 diabetes on structural changes and hormonal output of the adrenal cortex in male Wistar rats. J Diabetes Metab Disord 2013; 12: 9
  • 27 Astort F, Repetto EM, Martinez Calejman C, Cipelli JM, Sanchez R, Di Gruccio JM, Mercau M, Pignataro OP, Arias P, Cymeryng CB. High glucose-induced changes in steroid production in adrenal cells. Diabetes Metab Res Rev 2009; 25: 477-486
  • 28 van Haarst AD, Oitzl MS, de Kloet ER. Facilitation of feedback inhibition through blockade of glucocorticoid receptors in the hippocampus. Neurochem Res 1997; 22: 1323-1328
  • 29 Kotelevtsev Y, Holmes MC, Burchell A, Houston PM, Schmoll D, Jamieson P, Best R, Brown R, Edwards CR, Seckl JR, Mullins JJ. 11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. Proc Natl Acad Sci USA 1997; 94: 14924-14929
  • 30 Lamberts SW, Koper JW, de Jong FH. Long-term treatment with RU486 and glucocorticoid receptor resistance: theoretical and therapeutic implications. Trends Endocrinol Metab 1992; 3: 199-204
  • 31 van Haarst AD, Oitzl MS, Workel JO, de Kloet ER. Chronic brain glucocorticoid receptor blockade enhances the rise in circadian and stress-induced pituitary-adrenal activity. Endocrinology 1996; 137: 4935-4943
  • 32 Reul JM, Probst JC, Skutella T, Hirschmann M, Stec IS, Montkowski A, Landgraf R, Holsboer F. Increased stress-induced adrenocorticotropin response after long-term intracerebroventricular treatment of rats with antisense mineralocorticoid receptor oligodeoxynucleotides. Neuroendocrinology 1997; 65: 189-199
  • 33 Rosmond R, Dallman MF, Bjorntorp P. Stress-related cortisol secretion in men: relationships with abdominal obesity and endocrine, metabolic and hemodynamic abnormalities. J Clin Endocrinol Metab 1998; 83: 1853-1859
  • 34 Rosmond R, Chagnon YC, Holm G, Chagnon M, Perusse L, Lindell K, Carlsson B, Bouchard C, Bjorntorp P. A glucocorticoid receptor gene marker is associated with abdominal obesity, leptin, and dysregulation of the hypothalamic-pituitary-adrenal axis. Obesity Res 2000; 8: 211-218
  • 35 Akana SF, Dallman MF, Bradbury MJ, Scribner KA, Strack AM, Walker CD. Feedback and facilitation in the adrenocortical system: unmasking facilitation by partial inhibition of the glucocorticoid response to prior stress. Endocrinology 1992; 131: 57-68
  • 36 Akana SF, Scribner KA, Bradbury MJ, Strack AM, Walker CD, Dallman MF. Feedback sensitivity of the rat hypothalamo-pituitary-adrenal axis and its capacity to adjust to exogenous corticosterone. Endocrinology 1992; 131: 585-594
  • 37 Gallo PV, Weinberg J. Corticosterone rhythmicity in the rat: interactive effects of dietary restriction and schedule of feeding. J Nutr 1981; 111: 208-218
  • 38 Marissal-Arvy N, Gaumont A, Langlois A, Dabertrand F, Bouchecareilh M, Tridon C, Mormede P. Strain differences in hypothalamic pituitary adrenocortical axis function and adipogenic effects of corticosterone in rats. J Endocrinol 2007; 195: 473-484