Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-9q27g Total loading time: 0 Render date: 2024-07-23T10:51:55.144Z Has data issue: false hasContentIssue false

22 - Antenatal glucocorticoids and programming of neuroendocrine function and behavior

from Section 5 - Follow-up

Published online by Cambridge University Press:  01 March 2011

Hugo Lagercrantz
Affiliation:
Karolinska Institutet, Stockholm
M. A. Hanson
Affiliation:
Southampton General Hospital
Laura R. Ment
Affiliation:
Yale University, Connecticut
Donald M. Peebles
Affiliation:
University College London
Get access

Summary

Introduction

Glucocorticoids (cortisol in humans and most mammals, and corticosterone in rats, mice, and other lower vertebrates) are essential for normal brain development. They exert a wide spectrum of effects in most regions of the developing brain, ranging from subcellular reorganization to neuron–neuron and neuron–glial interaction. However, sustained elevation in, or removal of these hormones from, the fetal brain is detrimental to these processes, and can permanently modify the structure and function of the brain (Charmandari et al., 2003).

The pioneering work of Liggins and Howie in the early 1970s led to the widespread use of synthetic glucocorticoids to treat fetuses at risk of preterm delivery (Liggins & Howie, 1972). Preterm delivery occurs in approximately 7% of pregnancies in North America and results in approximately 75% of all neonatal deaths. In such cases, antenatal synthetic glucocorticoid therapy, administered between 24 and 34 weeks' gestation is highly effective in reducing the frequency of respiratory complications and perinatal death (National Institutes of Health [NIH] Consensus Panel, 1995). In 1994, the NIH endorsed the use of maternal synthetic glucocorticoid therapy during pregnancy (NIH Consensus Panel, 1995) and a similar approach was adopted around the world. Due to difficulty in predicting preterm birth, many women received repeat courses of synthetic glucocorticoids. Indeed, surveys in the late 1990s revealed that repeated dosing had become common practice in the late 1990s, with some women receiving up to 11 repeat courses (Brocklehurst et al., 1999).

Type
Chapter
Information
The Newborn Brain
Neuroscience and Clinical Applications
, pp. 361 - 376
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Andrews, M. H., Kostaki, A., Setiawan, E., et al. (2004). Developmental regulation of the 5-HT7 serotonin receptor and transcription factor NGFI-A in the fetal guinea-pig limbic system: Influence of GCs. Journal of Physiology, 555, 659–70.CrossRefGoogle ScholarPubMed
Bakker, J. M., Schmidt, E. D., Kroes, H., et al. (1995). Effects of short-term dexamethasone treatment during pregnancy on the development of the immune system and the hypothalamo-pituitary adrenal axis in the rat. Journal of Neuroimmunology, 63, 183–91.CrossRefGoogle ScholarPubMed
Bremner, J. D., Randall, P., Scott, T. M., et al. (1995). MRI-based measurement of hippocampal volume in patients with combat-related posttraumatic stress disorder. American Journal of Psychiatry, 152, 973–81.Google ScholarPubMed
Brocklehurst, P., Gates, S., McKenzie-McHarg, K., et al. (1999). Are we prescribing multiple courses of antenatal corticosteroids? A survey of practice in the UK. British Journal of Obstetrics and Gynaecology, 106, 977–9.CrossRefGoogle ScholarPubMed
Burdge, G. C., Slater-Jefferies, J., Torrens, C., et al. (2007). Dietary protein restriction of pregnant rats in the F0 generation induces altered methylation of hepatic gene promoters in the adult male offspring in the F1 and F2 generations. British Journal of Nutrition, 97, 435–9.CrossRefGoogle ScholarPubMed
Burlet, G., Fernette, B., Blanchard, S., et al. (2005). Antenatal glucocorticoids blunt the functioning of the hypothalamic-pituitary-adrenal axis of neonates and disturb some behaviors in juveniles. Neuroscience, 133, 221–30.CrossRefGoogle ScholarPubMed
Caldji, C., Tannenbaum, B., Sharma, S., et al. (1998). Maternal care during infancy regulates the development of neural systems mediating the expression of fearfulness in the rat. Proceedings of the National Academy of Sciences of the USA, 95, 5335–40.CrossRefGoogle ScholarPubMed
Challis, J. R. G., Matthews, S. G., Gibb, W., et al. (2000). Endocrine and paracrine regulation of birth at term and preterm. Endocrine Reviews, 21, 514–50.Google ScholarPubMed
Charmandari, E., Kino, T., Souvatzoglou, E., et al. (2003). Pediatric stress: Hormonal mediators and human development. Hormone Research, 59, 161–79.CrossRefGoogle ScholarPubMed
Coe, C. L. & Lubach, G. R. (2005). Developmental consequences of antenatal dexamethasone treatment in nonhuman primates. Neuroscience and Biobehavioral Reviews, 29, 227–35.CrossRefGoogle ScholarPubMed
Coe, C. L., Kramer, M., Czeh, B., et al. (2003). Prenatal stress diminishes neurogenesis in the dentate gyrus of juvenile rhesus monkeys. Biological Psychiatry, 54, 1025–34.CrossRefGoogle ScholarPubMed
,Committee on Obstetric Practice. (2002). ACOG committee opinion: Antenatal corticosteroid therapy for fetal maturation. Obstetrics and Gynecology, 99, 871–3.Google Scholar
Crowther, C. A., Haslam, R. R., Hiller, J. E., et al. (2006). Neonatal respiratory distress syndrome after repeat exposure to antenatal corticosteroids: a randomised controlled trial. Lancet, 367, 1913–19.CrossRefGoogle ScholarPubMed
Dalziel, S. R., Lim, V. K., Lambert, A., et al. (2005). Antenatal exposure to betamethasone: Psychological functioning and health related quality of life 31 years after inclusion in randomised controlled trial. British Medical Journal, 331, 665.CrossRefGoogle ScholarPubMed
Davis, E. P., Townsend, E. L., Gunnar, M. R., et al. (2006). Antenatal betamethasone treatment has a persisting influence on infant HPA axis regulation. Journal of Perinatology, 26, 147–53.CrossRefGoogle Scholar
Kloet, E. R., Joels, M. & Holsboer, F. (2005). Stress and the brain: From adaptation to disease. Nature Reviews Neuroscience, 6, 463–75.CrossRefGoogle ScholarPubMed
Vries, A., Holmes, M. C., Heijnis, A., et al. (2007). Prenatal dexamethasone exposure induces changes in nonhuman primate offspring cardiometabolic and hypothalamic-pituitary-adrenal axis function. Journal of Clinical Investigation, 117, 1058–67.CrossRefGoogle ScholarPubMed
Dean, F., Yu, C., Lingas, R. I., et al. (2001). Prenatal glucocorticoid modifies hypothalamo-pituitary-adrenal regulation in prepubertal guinea pigs. Neuroendocrinology, 73, 194–202.CrossRefGoogle ScholarPubMed
Dobbing, J. & Sands, J. (1979). Comparative aspects of the brain growth spurt. Early Human Development, 3, 79–83.CrossRefGoogle ScholarPubMed
Drake, A. J., Walker, B. R. & Seckl, J. R. (2005). Intergenerational consequences of fetal programming by in utero exposure to glucocorticoids in rats. American Journal of Physiology, Regulatory, Integrative and Comparative Physiology, 288, R34–8.CrossRefGoogle ScholarPubMed
Dunn, E., Owen, D., Kostaki, A., et al. (2005). Prenatal glucocorticoid exposure affects reproductive capacity and adrenocortical function in mature female guinea pig offspring. Pediatric Research 58, 0–800.Google Scholar
Erdeljan, P., Andrews, M. H., MacDonald, J. F., et al. (2005). Glucocorticoids and serotonin alter glucocorticoid receptor mRNA levels in fetal guinea-pig hippocampal neurons, in vitro. Reproduction, Fertility and Development, 17, 743–9.CrossRefGoogle ScholarPubMed
Francis, D., Diorio, J., Liu, D., et al. (1999). Nongenomic transmission across generations of maternal behavior and stress responses in the rat. Science, 286, 1155–8.CrossRefGoogle ScholarPubMed
French, N. P., Hagan, R., Evans, S. F., et al. (2004). Repeated antenatal corticosteroids: Effects on cerebral palsy and childhood behavior. American Journal of Obstetrics and Gynaecology, 190, 588–95.CrossRefGoogle ScholarPubMed
Fries, E., Hesse, J., Hellhammer, J., et al. (2005). A new view on hypocortisolism. Psychoneuroendocrinology, 30, 1010–16.CrossRefGoogle ScholarPubMed
Hauser, J., Dettling-Artho, A., Pilloud, S., et al. (2007). Effects of prenatal dexamethasone treatment on postnatal physical, endocrine, and social development in the common marmoset monkey. Endocrinology, 148, 1813–22.CrossRefGoogle ScholarPubMed
Herman, J. P., Prewitt, C. M. & Cullinan, W. E. (1996). Neuronal circuit regulation of the hypothalamo-pituitary-adrenocortical stress axis. Critical Reviews in Neurobiology, 10, 371–94.CrossRefGoogle ScholarPubMed
Hirvikoski, T., Nordenstrom, A., Lindholm, T., et al. (2007). Cognitive functions in children at risk for congenital adrenal hyperplasia treated prenatally with dexamethasone. Journal of Clinical Endocrinology and Metabolism, 92, 542–8.CrossRefGoogle ScholarPubMed
John, C. D., Theogaraj, E., Christian, H. C., et al. (2006). Time-specific effects of perinatal glucocorticoid treatment on anterior pituitary morphology, annexin 1 expression and adrenocorticotrophic hormone secretion in the adult female rat. Journal of Neuroendocrinology, 18, 949–59.CrossRefGoogle ScholarPubMed
Kliewer, S. A., Moore, J. T., Wade, L., et al. (1998). An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell, 92, 73–82.CrossRefGoogle ScholarPubMed
Levitt, N. S., Lindsay, R. S., Holmes, M. C., et al. (1996). Dexamethasone in the last week of pregnancy attenuates hippocampal glucocorticoid receptor gene expression and elevates blood pressure in the adult offspring in the rat. Neuroendocrinology, 64, 412–18.CrossRefGoogle ScholarPubMed
Liggins, G. C. & Howie, R. N. (1972). A controlled trial of antepartum glucocorticoid treatment for prevention of the respiratory distress syndrome in premature infants. Pediatrics, 50, 515–25.Google ScholarPubMed
Lillycrop, K. A., Phillips, E. S., Jackson, A. A., et al. (2005). Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. Journal of Nutrition, 135, 1382–6.CrossRefGoogle ScholarPubMed
Liu, D., Diorio, J., Tannenbaum, B., et al. (1997). Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science, 277, 1659–62.CrossRefGoogle ScholarPubMed
Liu, L., Li, A. & Matthews, S. G. (2001). Maternal glucocorticoid treatment programs HPA regulation in adult offspring: Sex-specific effects. American Journal of Physiology Endocrinology and Metabolism, 280, E729–39.CrossRefGoogle ScholarPubMed
Lupien, S. J. & McEwen, B. S. (1997). The acute effects of corticosteroids on cognition: Integration of animal and human model studies. Brain Research Brain Research Reviews, 24, 1–27.CrossRefGoogle ScholarPubMed
Ma, X. H., Wu, W. X. & Nathanielsz, P. W. (2003). Gestation-related and betamethasone-induced changes in 11beta-hydroxysteroid dehydrogenase types 1 and 2 in the baboon placenta. American Journal of Obstetrics and Gynaecology, 188, 13–21.CrossRefGoogle ScholarPubMed
McCabe, L., Marash, D., Li, A., et al. (2001). Repeated antenatal glucocorticoid treatment decreases hypothalamic corticotropin releasing hormone mRNA but not corticosteroid receptor mRNA expression in the fetal guinea-pig brain. Journal of Neuroendocrinology, 13, 425–31.CrossRefGoogle Scholar
Meaney, M. J., Diorio, J., Francis, D., et al. (2000). Postnatal handling increases the expression of cAMP-inducible transcription factors in the rat hippocampus: The effects of thyroid hormones and serotonin. Journal of Neuroscience, 20, 3926–35.CrossRefGoogle ScholarPubMed
Meaney, M. J., Brake, W. & Gratton, A. (2002). Environmental regulation of the development of mesolimbic dopamine systems: A neurobiological mechanism for vulnerability to drug abuse?Psychoneuroendocrinology, 27, 127–38.CrossRefGoogle ScholarPubMed
Muneoka, K., Mikuni, M., Ogawa, T., et al. (1997). Prenatal dexamethasone exposure alters brain monoamine metabolism and adrenocortical response in rat offspring. American Journal of Physiology, 273, R1669–75.Google ScholarPubMed
,National Institutes of Health (NIH) Consensus Panel. (1995). Effect of corticosteroids for fetal maturation on perinatal outcomes. NIH consensus development panel on the effect of corticosteroids for fetal maturation on perinatal outcomes. JAMA: The Journal of American Medical Association, 273, 413–18.CrossRefGoogle Scholar
Noorlander, C. W., Graan, P. N., Middeldorp, J., et al. (2006). Ontogeny of hippocampal corticosteroid receptors: Effects of antenatal glucocorticoids in human and mouse. Journal of Comparative Neurology, 499, 924–32.CrossRefGoogle Scholar
O'Connor, T. G., Heron, J., Golding, J., et al. (2003). Maternal antenatal anxiety and behavioural/emotional problems in children: a test of a programming hypothesis. Journal of Child Psychology and Psychiatry, 44, 1025–36.CrossRefGoogle ScholarPubMed
O'Regan, D., Kenyon, C. J., Seckl, J. R., et al. (2004). Glucocorticoid exposure in late gestation in the rat permanently programs gender-specific differences in adult cardiovascular and metabolic physiology. American Journal of Physiology Endocrinology and Metabolism, 287, E863–70.CrossRefGoogle ScholarPubMed
Owen, D. & Matthews, S. G. (2003). Glucocorticoids and sex-dependent development of brain glucocorticoid and mineralocorticoid receptors. Endocrinology, 144, 2775–84.CrossRefGoogle ScholarPubMed
Owen, D. & Matthews, S. G. (2007a). Prenatal glucocorticoid exposure alters hypothalamic-pituitary-adrenal function in juvenile guinea pigs. Journal of Neuroendocrinology, 19, 172–80.CrossRefGoogle ScholarPubMed
Owen, D. & Matthews, S. G. (2007b). Repeated maternal glucocorticoid treatment affects activity and hippocampal NMDA receptor expression in juvenile guinea pigs. Journal of Physiology, 578, 249–57.CrossRefGoogle ScholarPubMed
Petropoulos, S., Kalabis, G. M., Gibb, W., et al. (2007). Functional changes of mouse placental multidrug resistance phosphoglycoprotein (ABCB1) with advancing gestation and regulation by progesterone. Reproductive Sciences, 14, 321–8.CrossRefGoogle ScholarPubMed
Sapolsky, R. M. (1996). Why stress is bad for your brain. Science, 273, 749–50.CrossRefGoogle ScholarPubMed
Seckl, J. R. & Holmes, M. C. (2007). Mechanisms of disease: Glucocorticoids, their placental metabolism and fetal “programming” of adult pathophysiology. Nature Clinical Practice Endocrinology and Metabolism, 3, 479–88.CrossRefGoogle ScholarPubMed
Semont, A., Fache, M., Ouafik, L., et al. (1999). Effect of serotonin inhibition on glucocorticoid and mineralocorticoid expression in various brain structures. Neuroendocrinology, 69, 121–8.CrossRefGoogle ScholarPubMed
Setiawan, E., Jackson, M. F., Macdonald, J. F., et al. (2007). Effects of repeated prenatal glucocorticoid exposure on LTP in the juvenile guinea pig hippocampus. Journal of Physiology, 581, 1033–42.CrossRefGoogle ScholarPubMed
Shoener, J. A., Baig, R. & Page, K. C. (2006). Prenatal exposure to dexamethasone alters hippocampal drive on hypothalamic-pituitary-adrenal axis activity in adult male rats. American Journal of Physiology Regulatory, Integrative and Comparative Physiology, 290, R1366–73.CrossRefGoogle ScholarPubMed
Sloboda, D. M., Newnham, J. P. & Challis, J. R. (2000). Effects of repeated maternal betamethasone administration on growth and hypothalamic-pituitary-adrenal function of the ovine fetus at term. Journal of Endocrinology, 165, 79–91.CrossRefGoogle ScholarPubMed
Sloboda, D. M., Moss, T. J., Li, S., et al. (2007). Prenatal betamethasone exposure results in pituitary-adrenal hyporesponsiveness in adult sheep. American Journal of Physiology Endocrinology and Metabolism, 292, E61–70.CrossRefGoogle ScholarPubMed
Slotkin, T. A., Lappi, S. E., McCook, E. C., et al. (1992). Glucocorticoids and the development of neuronal function: Effects of prenatal dexamethasone exposure on central noradrenergic activity. Biology of the Neonate, 61, 326–36.CrossRefGoogle ScholarPubMed
Slotkin, T. A., Barnes, G. A., McCook, E. C., et al. (1996). Programming of brainstem serotonin transporter development by prenatal glucocorticoids. Brain Research Development, 93, 155–61.CrossRefGoogle ScholarPubMed
Stein, M. B., Koverola, C., Hanna, C., et al. (1997). Hippocampal volume in women victimized by childhood sexual abuse. Psychological Medicine, 27, 951–9.CrossRefGoogle ScholarPubMed
Stojanoski, M. M., Nestorovic, N., Negic, N., et al. (2006). The pituitary-adrenal axis of fetal rats after maternal dexamethasone treatment. Anatomy and Embryology, 211, 61–9.CrossRefGoogle ScholarPubMed
Sun, M., Kingdom, J., Baczyk, D., et al. (2006). Expression of the multidrug resistance P-glycoprotein, (ABCB1 glycoprotein) in the human placenta decreases with advancing gestation. Placenta, 27, 602–9.CrossRefGoogle ScholarPubMed
Szyf, M. (2007). The dynamic epigenome and its implications in toxicology. Toxicological Sciences, 100, 7–23.CrossRefGoogle ScholarPubMed
Theogaraj, E., John, C. D., Christian, H. C., et al. (2005). Perinatal glucocorticoid treatment produces molecular, functional, and morphological changes in the anterior pituitary gland of the adult male rat. Endocrinology, 146, 4804–13.CrossRefGoogle ScholarPubMed
Uno, H., Lohmiller, L., Thieme, C., et al. (1990). Brain damage induced by prenatal exposure to dexamethasone in fetal rhesus macaques. I. Hippocampus. Brain Research Development Brain Research, 53, 157–67.CrossRefGoogle ScholarPubMed
Uno, H., Eisele, S., Sakai, A., et al. (1994). Neurotoxicity of glucocorticoids in the primate brain. Hormones and Behavior, 28, 336–48.CrossRefGoogle ScholarPubMed
Viau, V. (2002). Functional cross-talk between the hypothalamic-pituitary-gonadal and adrenal axes. Journal of Neuroendocrinology, 14, 506–13.CrossRefGoogle ScholarPubMed
Wan, S., Hao, R. & Sun, K. (2005). Repeated maternal dexamethasone treatments in late gestation increases 11beta-hydroxysteroid dehydrogenase type 1 expression in the hippocampus of the newborn rat. Neuroscience Letters, 382, 96–101.CrossRefGoogle ScholarPubMed
Weaver, I. C., Cervoni, N., Champagne, F. A., et al. (2004). Epigenetic programming by maternal behavior. Nature Neuroscience, 7, 847–54.CrossRefGoogle ScholarPubMed
Weaver, I. C., D'Alessio, A. C., Brown, S. E., et al. (2007). The transcription factor nerve growth factor-inducible protein a mediates epigenetic programming: Altering epigenetic marks by immediate-early genes. Journal of Neuroscience, 27, 1756–68.CrossRefGoogle ScholarPubMed
Welberg, L. A., Seckl, J. R. & Holmes, M. C. (2001). Prenatal glucocorticoid programming of brain corticosteroid receptors and corticotrophin-releasing hormone: Possible implications for behaviour. Neuroscience, 104, 71–9.CrossRefGoogle ScholarPubMed
Yu, L., Cohen, D., Piekarz, R. L., et al. (1993). Three distinct nuclear protein binding sites in the promoter of the murine multidrug resistance mdr1b gene. Journal of Biological Chemistry, 268, 7520–6.Google ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×