Horm Metab Res 2008; 40(4): 257-261
DOI: 10.1055/s-2008-1058064
Animals, Clinical

© Georg Thieme Verlag KG Stuttgart · New York

Tissue-specific Programming Expression of Glucocorticoid Receptors and 11β-HSDs by Maternal Perinatal Undernutrition in the HPA Axis of Adult Male Rats

I. Dutriez-Casteloot 1 [*] , C. Breton 1 [*] , B. Coupé 1 , O. Hawchar 1 , M. Enache 1 , A. Dickes-Coopman 1 , Y. de. Keyzer 2 , S. Deloof 1 , J. Lesage 1 , D. Vieau 1
  • 1Neurosciences et Neurophysiologie Adaptatives, Equipe Stress Périnatal, Université de Lille I, Villeneuve d’Ascq, France
  • 2Institut Cochin, Paris, France
Further Information

Publication History

received 01.06.2007

accepted 10.09.2007

Publication Date:
03 March 2008 (online)

Abstract

Maternal undernutrition leads to intrauterine growth retardation and predisposes to the development of pathologies in adulthood. The hypothalamo-pituitary-adrenal axis is a major target of early-life programming. We showed previously that perinatal maternal 50% food restriction leads to hypothalamo-pituitary-adrenal axis hyperactivity and disturbs glucocorticoid feedback in adult male rats. To try to better understand these alterations, we studied several factors involved in corticosterone sensitivity. We showed that unlike the restricted expression of 11β-HSD2 mRNA, the 11β-HSD1, glucocorticoid, and mineralocorticoid receptor genes are widely distributed in rat. In contrast to the hypothalamus, we confirmed that maternal undernutrition modulates hippocampal corticosterone receptor balance and leads to increased 11β-HSD1 gene expression. In the pituitary, rats exhibited a huge increase in both mRNA and mineralocorticoid receptor binding capacities as well as decreased 11β-HSD1/11β-HSD2 gene expression. Using in situ hybridization, we showed that the mineralocorticoid receptor gene was expressed in rat corticotroph cells and by other adenopituitary cells. In the adrenal gland, maternal food restriction decreased 11β-HSD2 mRNA. This study demonstrated that maternal food restriction has both long-term and tissue-specific effects on gene expression of factors involved in glucocorticoid sensitivity and that it could contribute, via glucocorticoid excess, to the development of adult diseases.

References

  • 1 Barker DJP. In utero programming of chronic disease.  Clin Sci. 1998;  95 115-128
  • 2 Lesage J, Sebaai N, Leonhardt M, Dutriez-Casteloot I, Breton C, Deloof S, Vieau D. Perinatal maternal undernutrition programs the offspring hypothalamo-pituitary-adrenal axis in mammals.  Stress. 2006;  9 183-198
  • 3 O’Regan D, Kenyon JC, Seckl JR, Holmes MC. Glucocorticoid exposure in late gestation in the rat permanently programs gender-specific differences in adult cardiovascular and metabolic physiology.  Am J Physiol Endocrinol Metab. 2004;  287 863-870
  • 4 Plagemann A. ‘Fetal programming’ and ‘functional teratogenesis’: on epigenetic mechanisms and prevention of perinatally acquired lasting health risks.  J Perinat Med. 2004;  32 297-305
  • 5 Seckl JR. Prenatal glucocorticoids and long-term programming.  Eur J Endocrinology. 2004;  151 49-62
  • 6 Lemaire V, Koehl M, Le Moal M, Abrous DM. Prenatal stress products learning deficits associated with an inhibition of neurogenesis in the hippocampus.  Proc Natl Acad Sci USA. 2000;  97 11032-11037
  • 7 Matthews SG. Antenatal glucocorticoids and programming of the developing CNS.  Pediatr Res. 2000;  47 291-300
  • 8 Welberg LAM, Seckl JR, Holmes MC. Prenatal glucocorticoid programming of brain corticosteroid receptors and corticotrophin-releasing hormone: Possible implications for behaviour.  Neuroscience. 2001;  104 71-79
  • 9 Leonhardt M, Lesage J, Dufourny L, Dickes-Coopman A, Montel V, Dupouy JP. Perinatal maternal food restriction induces alterations in the hypothalamo-pituitary adrenal axis activity and in plasma corticosterone binding globulin capacity of weaning rat pups.  Neuroendocrinology. 2002;  75 45-54
  • 10 Sebaai N, Lesage J, Vieau D, Alaoui A, Dupouy JP, Deloof S. Altered control of the hypothalamo-pituitary adrenal axis in perinatally malnourished adult male rats: effects of a dehydration period.  Neuroendocrinology. 2002;  74 243-253
  • 11 Sebaai N, Lesage J, Breton C, Vieau D, Deloof S. Perinatal food deprivation induces marked alterations of the hypothalamo-pituitary-adrenal axis in 8 months male rats both under basal conditions and after a dehydration period.  Neuroendocrinology. 2004;  79 163-173
  • 12 Buckingham JC. Glucocorticoids: exemplars of multi-tasking.  Br J Pharmacol. 2006;  147 ((Suppl 1)) 258-268
  • 13 Breton C, Pechoux C, Morel Z, Zingg HH. Oxytocin receptor messenger ribonucleic acid: characterization, regulation, and cellular localization in the rat pituitary gland.  Endocrinology. 1995;  136 2928-2936
  • 14 Dutriez-Casteloot I, Montel V, Croix D, Laborie C, Van Camp G, Beauvillain JC, Dupouy JP. Activities of the pituitary-adrenal and gonadal axes during the estrous cycle in adult female rats prenatally exposed to morphine.  Brain Research. 2001;  902 66-73
  • 15 Drouin J, Chamberland M, Charron J, Jeannotte L, Nemer M. Structure of the rat proopiomelanocortin (POMC) gene.  FEBS Lett. 1985;  193 54-58
  • 16 Seckl JR. 11β-hydroxysteroid dehydrogenases: changing glucocorticoid action.  Current Opinion in Pharmacology. 2004;  4 597-602
  • 17 Naray-Fejes-Toth A, Fejes-Toth G. Extranuclear localization of endogenous 11β-hydroxysteroid dehydrogenase-2 in aldosterone target cells.  Endocrinology. 1998;  139 2955-2959
  • 18 Roland BL, Funder JW. Localization of 11β-hydroxysteroid dehydrogenase type 2 in rat tissues: in situ studies.  Endocrinology. 1996;  137 1123-1128
  • 19 Brown RW, Diaz R, Robson AC, Kotelevtsev YV, Mullins JJ, Kaufman MH, Seckl JR. The ontogeny of 11 beta-hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development.  Endocrinology. 1996;  137 794-797
  • 20 Oitzl MS, Haarst AD Van, Sutanto W, Kloet ER De. Corticosterone, brain mineralocorticoid receptors (MRs) and the activity of the hypothalamic-pituitary-adenal (HPA) axis: the Lewis rat as an example of increased central MR capacity and a hyporesponsive HPA axis.  Psychoneuroendocrinology. 1995;  20 655-675
  • 21 Rabbitt EH, Gittoes NJ, Stewart PM, Hewison M. 11beta-hydroxysteroid dehydrogenases, cell proliferation and malignancy.  J Steroid Biochem Mol Biol. 2003;  85 415-421
  • 22 Shimojo M, Condon J, Whorwood CB, Stewart PM. Adrenal 11b-hydroxysteroid dehydrogenase.  Endoc Res. 1996;  22 771-780
  • 23 Cole TJ, Blendy JA, Monghan AP, Krieglstein K, Schmid W, Aguzzi A, Fantuzzi G, Hummler E, Unsicker K, Schultz G. Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation.  Genes Dev. 1995;  9 1608-1621
  • 24 Ricketts ML, Verhaeg JM, Bujalska I, Howie AJ, Rainey WE, Stewart PM. Immunohistochemical localization of type 1 11β-hydroxysteroid dehydrogenase in human tissues.  J Clin Endocrinol Metab. 1998;  83 1325-1335
  • 25 Frey FJ. Methylation of CpG islands: potential relevance for hypertension and kidney diseases.  Nephrol Dial Transplant. 2005;  20 868-869
  • 26 Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney MJ. Epigenetic programming by maternal behaviour.  Nat Neurosci. 2004;  7 847-854

1 These authors contributed equally to this work.

Correspondence

Dr. D. Vieau

Neurosciences et Physiologie Adaptatives

UPRES EA4052

Equipe Stress Périnataux SN4 Université de Lille I

59655 Villeneuve d'Ascq

France

Phone: +33/3/2043 43 68

Fax: +33/3/2033 63 49

Email: Didier.Vieau@univ-lille1.fr

    >