Skip to main content
Top
Published in: Seminars in Immunopathology 6/2016

01-11-2016 | Review

Antenatal endogenous and exogenous glucocorticoids and their impact on immune ontogeny and long-term immunity

Authors: María Emilia Solano, Megan C. Holmes, Paul R. Mittelstadt, Karen E. Chapman, Eva Tolosa

Published in: Seminars in Immunopathology | Issue 6/2016

Login to get access

Abstract

Endogenous levels of glucocorticoids rise during pregnancy to warrant development and maturation of the fetal organs close to birth. However, during most of the gestation, the fetus is protected from excessive biologically active endogenous glucocorticoids by placental and fetal expression of 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2). Maternal stress, which may overwhelm placental 11β-HSD2 activity with high glucocorticoid levels, or administration of synthetic glucocorticoids to improve the survival chances of the premature newborn, are associated to postnatal increased risk for immune diseases. Fetal exposure to excessive glucocorticoids may underlie this altered postnatal immunity. Here, we revise the role that placental and fetal 11β-HSD2, fetal glucocorticoid exposure, and programming of the offspring’s the hypothalamic-pituitary-adrenal (HPA) axis play on concerted steps in immune fetal development. We could identify gaps in knowledge about glucocorticoid-induced programming of immune diseases. Finally, based on current evidence about glucocorticoid and HPA axis-mediated immune regulation, we hypothesize on mechanisms that could drive the enhanced risk for atopies, infections, and type I diabetes in offspring that were prenatally exposed to glucocorticoids.
Literature
2.
go back to reference Douglas AJ (2010) Mother-offspring dialogue in early pregnancy: impact of adverse environment on pregnancy maintenance and neurobiology. Prog Neuropsychopharmacol Biol Psychiatry 35(5):1167–1177PubMedCrossRef Douglas AJ (2010) Mother-offspring dialogue in early pregnancy: impact of adverse environment on pregnancy maintenance and neurobiology. Prog Neuropsychopharmacol Biol Psychiatry 35(5):1167–1177PubMedCrossRef
3.
go back to reference Jung C, Ho JT, Torpy DJ, Rogers A, Doogue M, Lewis JG, Czajko RJ, Inder WJ (2011) A longitudinal study of plasma and urinary cortisol in pregnancy and postpartum. J Clin Endocrinol Metab 96(5):1533–1540PubMedCrossRef Jung C, Ho JT, Torpy DJ, Rogers A, Doogue M, Lewis JG, Czajko RJ, Inder WJ (2011) A longitudinal study of plasma and urinary cortisol in pregnancy and postpartum. J Clin Endocrinol Metab 96(5):1533–1540PubMedCrossRef
4.
go back to reference Wyrwoll CS, Seckl JR, Holmes MC (2009) Altered placental function of 11beta-hydroxysteroid dehydrogenase 2 knockout mice. Endocrinology 150(3):1287–1293PubMedCrossRef Wyrwoll CS, Seckl JR, Holmes MC (2009) Altered placental function of 11beta-hydroxysteroid dehydrogenase 2 knockout mice. Endocrinology 150(3):1287–1293PubMedCrossRef
5.
go back to reference Holmes MC, Abrahamsen CT, French KL, Paterson JM, Mullins JJ, Seckl JR (2006) The mother or the fetus? 11beta-hydroxysteroid dehydrogenase type 2 null mice provide evidence for direct fetal programming of behavior by endogenous glucocorticoids. J Neurosci 26(14):3840–3844PubMedCrossRef Holmes MC, Abrahamsen CT, French KL, Paterson JM, Mullins JJ, Seckl JR (2006) The mother or the fetus? 11beta-hydroxysteroid dehydrogenase type 2 null mice provide evidence for direct fetal programming of behavior by endogenous glucocorticoids. J Neurosci 26(14):3840–3844PubMedCrossRef
9.
go back to reference Duthie L, Reynolds RM (2013) Changes in the maternal hypothalamic-pituitary-adrenal axis in pregnancy and postpartum: influences on maternal and fetal outcomes. Neuroendocrinology 98(2):106–115PubMedCrossRef Duthie L, Reynolds RM (2013) Changes in the maternal hypothalamic-pituitary-adrenal axis in pregnancy and postpartum: influences on maternal and fetal outcomes. Neuroendocrinology 98(2):106–115PubMedCrossRef
10.
go back to reference Lindsay JR, Nieman LK (2005) The hypothalamic-pituitary-adrenal axis in pregnancy: challenges in disease detection and treatment. Endocr Rev 26(6):775–799PubMedCrossRef Lindsay JR, Nieman LK (2005) The hypothalamic-pituitary-adrenal axis in pregnancy: challenges in disease detection and treatment. Endocr Rev 26(6):775–799PubMedCrossRef
11.
go back to reference Reis FM, Fadalti M, Florio P, Petraglia F (1999) Putative role of placental corticotropin-releasing factor in the mechanisms of human parturition. J Soc Gynecol Investig 6(3):109–119PubMedCrossRef Reis FM, Fadalti M, Florio P, Petraglia F (1999) Putative role of placental corticotropin-releasing factor in the mechanisms of human parturition. J Soc Gynecol Investig 6(3):109–119PubMedCrossRef
12.
go back to reference Petraglia F, Potter E, Cameron VA, Sutton S, Behan DP, Woods RJ, Sawchenko PE, Lowry PJ, Vale W (1993) Corticotropin-releasing factor-binding protein is produced by human placenta and intrauterine tissues. J Clin Endocrinol Metab 77(4):919–924PubMed Petraglia F, Potter E, Cameron VA, Sutton S, Behan DP, Woods RJ, Sawchenko PE, Lowry PJ, Vale W (1993) Corticotropin-releasing factor-binding protein is produced by human placenta and intrauterine tissues. J Clin Endocrinol Metab 77(4):919–924PubMed
13.
go back to reference Robinson BG, Emanuel RL, Frim DM, Majzoub JA (1988) Glucocorticoid stimulates expression of corticotropin-releasing hormone gene in human placenta. Proc Natl Acad Sci U S A 85(14):5244–5248PubMedPubMedCentralCrossRef Robinson BG, Emanuel RL, Frim DM, Majzoub JA (1988) Glucocorticoid stimulates expression of corticotropin-releasing hormone gene in human placenta. Proc Natl Acad Sci U S A 85(14):5244–5248PubMedPubMedCentralCrossRef
14.
go back to reference Mastorakos G, Ilias I (2003) Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. Ann N Y Acad Sci 997:136–149PubMedCrossRef Mastorakos G, Ilias I (2003) Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. Ann N Y Acad Sci 997:136–149PubMedCrossRef
16.
go back to reference Douglas AJ, Brunton PJ, Bosch OJ, Russell JA, Neumann ID (2003) Neuroendocrine responses to stress in mice: hyporesponsiveness in pregnancy and parturition. Endocrinology 144(12):5268–5276PubMedCrossRef Douglas AJ, Brunton PJ, Bosch OJ, Russell JA, Neumann ID (2003) Neuroendocrine responses to stress in mice: hyporesponsiveness in pregnancy and parturition. Endocrinology 144(12):5268–5276PubMedCrossRef
17.
go back to reference Mizoguchi Y, Yamaguchi H, Aoki F, Enami J, Sakai S (1997) Corticosterone is required for the prolactin receptor gene expression in the late pregnant mouse mammary gland. Mol Cell Endocrinol 132(1–2):177–183PubMedCrossRef Mizoguchi Y, Yamaguchi H, Aoki F, Enami J, Sakai S (1997) Corticosterone is required for the prolactin receptor gene expression in the late pregnant mouse mammary gland. Mol Cell Endocrinol 132(1–2):177–183PubMedCrossRef
18.
go back to reference Johnstone HA, Wigger A, Douglas AJ, Neumann ID, Landgraf R, Seckl JR, Russell JA (2000) Attenuation of hypothalamic-pituitary-adrenal axis stress responses in late pregnancy: changes in feedforward and feedback mechanisms. J Neuroendocrinol 12(8):811–822PubMedCrossRef Johnstone HA, Wigger A, Douglas AJ, Neumann ID, Landgraf R, Seckl JR, Russell JA (2000) Attenuation of hypothalamic-pituitary-adrenal axis stress responses in late pregnancy: changes in feedforward and feedback mechanisms. J Neuroendocrinol 12(8):811–822PubMedCrossRef
19.
go back to reference Fowden AL, Forhead AJ (2015) Glucocorticoids as regulatory signals during intrauterine development. Exp Physiol 100(12):1477–1487PubMedCrossRef Fowden AL, Forhead AJ (2015) Glucocorticoids as regulatory signals during intrauterine development. Exp Physiol 100(12):1477–1487PubMedCrossRef
20.
go back to reference Fowden AL, Li J, Forhead AJ (1998) Glucocorticoids and the preparation for life after birth: are there long-term consequences of the life insurance? Proc Nutr Soc 57(1):113–122PubMedCrossRef Fowden AL, Li J, Forhead AJ (1998) Glucocorticoids and the preparation for life after birth: are there long-term consequences of the life insurance? Proc Nutr Soc 57(1):113–122PubMedCrossRef
21.
go back to reference Rog-Zielinska EA, Richardson RV, Denvir MA, Chapman KE (2014) Glucocorticoids and foetal heart maturation; implications for prematurity and foetal programming. J Mol Endocrinol 52(2):R125–135PubMedCrossRef Rog-Zielinska EA, Richardson RV, Denvir MA, Chapman KE (2014) Glucocorticoids and foetal heart maturation; implications for prematurity and foetal programming. J Mol Endocrinol 52(2):R125–135PubMedCrossRef
22.
go back to reference Vandevyver S, Dejager L, Tuckermann J, Libert C (2013) New insights into the anti-inflammatory mechanisms of glucocorticoids: an emerging role for glucocorticoid-receptor-mediated transactivation. Endocrinology 154(3):993–1007PubMedCrossRef Vandevyver S, Dejager L, Tuckermann J, Libert C (2013) New insights into the anti-inflammatory mechanisms of glucocorticoids: an emerging role for glucocorticoid-receptor-mediated transactivation. Endocrinology 154(3):993–1007PubMedCrossRef
23.
go back to reference Vandevyver S, Dejager L, Libert C (2012) On the trail of the glucocorticoid receptor: into the nucleus and back. Traffic 13(3):364–374PubMedCrossRef Vandevyver S, Dejager L, Libert C (2012) On the trail of the glucocorticoid receptor: into the nucleus and back. Traffic 13(3):364–374PubMedCrossRef
25.
go back to reference Coe CL, Lubach GR, Karaszewski JW (1999) Prenatal stress and immune recognition of self and nonself in the primate neonate. Biol Neonate 76(5):301–310PubMedCrossRef Coe CL, Lubach GR, Karaszewski JW (1999) Prenatal stress and immune recognition of self and nonself in the primate neonate. Biol Neonate 76(5):301–310PubMedCrossRef
26.
go back to reference Montano MM, Wang MH, Even MD, vom Saal FS (1991) Serum corticosterone in fetal mice: sex differences, circadian changes, and effect of maternal stress. Physiol Behav 50(2):323–329PubMedCrossRef Montano MM, Wang MH, Even MD, vom Saal FS (1991) Serum corticosterone in fetal mice: sex differences, circadian changes, and effect of maternal stress. Physiol Behav 50(2):323–329PubMedCrossRef
27.
go back to reference van Zon AA, Eling WM, Hermsen CC, Koekkoek AA (1982) Corticosterone regulation of the effector function of malarial immunity during pregnancy. Infect Immun 36(2):484–491PubMedPubMedCentral van Zon AA, Eling WM, Hermsen CC, Koekkoek AA (1982) Corticosterone regulation of the effector function of malarial immunity during pregnancy. Infect Immun 36(2):484–491PubMedPubMedCentral
28.
go back to reference Chang HY, Suh DI, Yang SI, Kang MJ, Lee SY, Lee E, Choi IA, Lee KS, Shin YJ, Shin YH, Kim YH, Kim KW, Ahn K, Won HS, Choi SJ, Oh SY, Kwon JY, Park HJ, Lee KJ, Jun JK, Yu HS, Lee SH, Jung BK, Kwon JW, Choi YK, Do N, Bae YJ, Kim H, Chang WS, Kim EJ, Lee JK, Hong SJ (2016) Prenatal maternal distress affects atopic dermatitis in offspring mediated by oxidative stress. J Allergy Clin Immunol. doi:10.1016/j.jaci.2016.01.020 PubMedCentral Chang HY, Suh DI, Yang SI, Kang MJ, Lee SY, Lee E, Choi IA, Lee KS, Shin YJ, Shin YH, Kim YH, Kim KW, Ahn K, Won HS, Choi SJ, Oh SY, Kwon JY, Park HJ, Lee KJ, Jun JK, Yu HS, Lee SH, Jung BK, Kwon JW, Choi YK, Do N, Bae YJ, Kim H, Chang WS, Kim EJ, Lee JK, Hong SJ (2016) Prenatal maternal distress affects atopic dermatitis in offspring mediated by oxidative stress. J Allergy Clin Immunol. doi:10.​1016/​j.​jaci.​2016.​01.​020 PubMedCentral
29.
go back to reference Mattos GE, Heinzmann JM, Norkowski S, Helbling JC, Minni AM, Moisan MP, Touma C (2013) Corticosteroid-binding globulin contributes to the neuroendocrine phenotype of mice selected for extremes in stress reactivity. J Endocrinol 219(3):217–229PubMedCrossRef Mattos GE, Heinzmann JM, Norkowski S, Helbling JC, Minni AM, Moisan MP, Touma C (2013) Corticosteroid-binding globulin contributes to the neuroendocrine phenotype of mice selected for extremes in stress reactivity. J Endocrinol 219(3):217–229PubMedCrossRef
31.
go back to reference Glavina-Durdov M, Springer O, Capkun V, Saratlija-Novakovic Z, Rozic D, Barle M (2003) The grade of acute thymus involution in neonates correlates with the duration of acute illness and with the percentage of lymphocytes in peripheral blood smear. Pathological study. Biol Neonate 83(4):229–234PubMedCrossRef Glavina-Durdov M, Springer O, Capkun V, Saratlija-Novakovic Z, Rozic D, Barle M (2003) The grade of acute thymus involution in neonates correlates with the duration of acute illness and with the percentage of lymphocytes in peripheral blood smear. Pathological study. Biol Neonate 83(4):229–234PubMedCrossRef
32.
go back to reference Chapman K, Holmes M, Seckl J (2013) 11beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. Physiol Rev 93(3):1139–1206PubMedPubMedCentralCrossRef Chapman K, Holmes M, Seckl J (2013) 11beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. Physiol Rev 93(3):1139–1206PubMedPubMedCentralCrossRef
33.
go back to reference Addison RS, Maguire DJ, Mortimer RH, Roberts MS, Cannell GR (1993) Pathway and kinetics of prednisolone metabolism in the human placenta. J Steroid Biochem Mol Biol 44(3):315–320PubMedCrossRef Addison RS, Maguire DJ, Mortimer RH, Roberts MS, Cannell GR (1993) Pathway and kinetics of prednisolone metabolism in the human placenta. J Steroid Biochem Mol Biol 44(3):315–320PubMedCrossRef
34.
go back to reference Gur C, Diav-Citrin O, Shechtman S, Arnon J, Ornoy A (2004) Pregnancy outcome after first trimester exposure to corticosteroids: a prospective controlled study. Reprod Toxicol 18(1):93–101PubMedCrossRef Gur C, Diav-Citrin O, Shechtman S, Arnon J, Ornoy A (2004) Pregnancy outcome after first trimester exposure to corticosteroids: a prospective controlled study. Reprod Toxicol 18(1):93–101PubMedCrossRef
35.
go back to reference Roberts D, Dalziel S (2006) Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev 3, CD004454 Roberts D, Dalziel S (2006) Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev 3, CD004454
36.
go back to reference Gyamfi-Bannerman C, Thom EA, Blackwell SC, Tita AT, Reddy UM, Saade GR, Rouse DJ, McKenna DS, Clark EA, Thorp JM Jr, Chien EK, Peaceman AM, Gibbs RS, Swamy GK, Norton ME, Casey BM, Caritis SN, Tolosa JE, Sorokin Y, VanDorsten JP, Jain L, NM-FMU Network (2016) Antenatal betamethasone for women at risk for late preterm delivery. N Engl J Med. doi:10.1056/NEJMoa1516783 Gyamfi-Bannerman C, Thom EA, Blackwell SC, Tita AT, Reddy UM, Saade GR, Rouse DJ, McKenna DS, Clark EA, Thorp JM Jr, Chien EK, Peaceman AM, Gibbs RS, Swamy GK, Norton ME, Casey BM, Caritis SN, Tolosa JE, Sorokin Y, VanDorsten JP, Jain L, NM-FMU Network (2016) Antenatal betamethasone for women at risk for late preterm delivery. N Engl J Med. doi:10.​1056/​NEJMoa1516783
38.
go back to reference Tegethoff M, Pryce C, Meinlschmidt G (2009) Effects of intrauterine exposure to synthetic glucocorticoids on fetal, newborn, and infant hypothalamic-pituitary-adrenal axis function in humans: a systematic review. Endocr Rev 30(7):753–789PubMedCrossRef Tegethoff M, Pryce C, Meinlschmidt G (2009) Effects of intrauterine exposure to synthetic glucocorticoids on fetal, newborn, and infant hypothalamic-pituitary-adrenal axis function in humans: a systematic review. Endocr Rev 30(7):753–789PubMedCrossRef
39.
go back to reference Alexander N, Rosenlocher F, Stalder T, Linke J, Distler W, Morgner J, Kirschbaum C (2012) Impact of antenatal synthetic glucocorticoid exposure on endocrine stress reactivity in term-born children. J Clin Endocrinol Metab 97(10):3538–3544. doi:10.1210/jc.2012-1970 PubMedCrossRef Alexander N, Rosenlocher F, Stalder T, Linke J, Distler W, Morgner J, Kirschbaum C (2012) Impact of antenatal synthetic glucocorticoid exposure on endocrine stress reactivity in term-born children. J Clin Endocrinol Metab 97(10):3538–3544. doi:10.​1210/​jc.​2012-1970 PubMedCrossRef
41.
go back to reference Diederich S, Eigendorff E, Burkhardt P, Quinkler M, Bumke-Vogt C, Rochel M, Seidelmann D, Esperling P, Oelkers W, Bahr V (2002) 11beta-hydroxysteroid dehydrogenase types 1 and 2: an important pharmacokinetic determinant for the activity of synthetic mineralo- and glucocorticoids. J Clin Endocrinol Metab 87(12):5695–5701. doi:10.1210/jc.2002-020970 PubMedCrossRef Diederich S, Eigendorff E, Burkhardt P, Quinkler M, Bumke-Vogt C, Rochel M, Seidelmann D, Esperling P, Oelkers W, Bahr V (2002) 11beta-hydroxysteroid dehydrogenase types 1 and 2: an important pharmacokinetic determinant for the activity of synthetic mineralo- and glucocorticoids. J Clin Endocrinol Metab 87(12):5695–5701. doi:10.​1210/​jc.​2002-020970 PubMedCrossRef
42.
go back to reference Kemp MW, Newnham JP, Challis JG, Jobe AH, Stock SJ (2016) The clinical use of corticosteroids in pregnancy. Hum Reprod Update 22(2):240–259PubMed Kemp MW, Newnham JP, Challis JG, Jobe AH, Stock SJ (2016) The clinical use of corticosteroids in pregnancy. Hum Reprod Update 22(2):240–259PubMed
43.
go back to reference Kajantie E, Raivio T, Janne OA, Hovi P, Dunkel L, Andersson S (2004) Circulating glucocorticoid bioactivity in the preterm newborn after antenatal betamethasone treatment. J Clin Endocrinol Metab 89(8):3999–4003PubMedCrossRef Kajantie E, Raivio T, Janne OA, Hovi P, Dunkel L, Andersson S (2004) Circulating glucocorticoid bioactivity in the preterm newborn after antenatal betamethasone treatment. J Clin Endocrinol Metab 89(8):3999–4003PubMedCrossRef
44.
go back to reference Fernandez-Balsells MM, Murad MH, Lane M, Lampropulos JF, Albuquerque F, Mullan RJ, Agrwal N, Elamin MB, Gallegos-Orozco JF, Wang AT, Erwin PJ, Bhasin S, Montori VM (2010) Clinical review 1: adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 95(6):2560–2575. doi:10.1210/jc.2009-2575 PubMedCrossRef Fernandez-Balsells MM, Murad MH, Lane M, Lampropulos JF, Albuquerque F, Mullan RJ, Agrwal N, Elamin MB, Gallegos-Orozco JF, Wang AT, Erwin PJ, Bhasin S, Montori VM (2010) Clinical review 1: adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 95(6):2560–2575. doi:10.​1210/​jc.​2009-2575 PubMedCrossRef
45.
go back to reference Alfaidy N, Gupta S, DeMarco C, Caniggia I, Challis JR (2002) Oxygen regulation of placental 11 beta-hydroxysteroid dehydrogenase 2: physiological and pathological implications. J Clin Endocrinol Metab 87(10):4797–4805PubMedCrossRef Alfaidy N, Gupta S, DeMarco C, Caniggia I, Challis JR (2002) Oxygen regulation of placental 11 beta-hydroxysteroid dehydrogenase 2: physiological and pathological implications. J Clin Endocrinol Metab 87(10):4797–4805PubMedCrossRef
46.
go back to reference Benediktsson R, Calder AA, Edwards CR, Seckl JR (1997) Placental 11 beta-hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure. Clin Endocrinol (Oxf) 46(2):161–166CrossRef Benediktsson R, Calder AA, Edwards CR, Seckl JR (1997) Placental 11 beta-hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure. Clin Endocrinol (Oxf) 46(2):161–166CrossRef
47.
go back to reference Cottrell EC, Holmes MC, Livingstone DE, Kenyon CJ, Seckl JR (2012) Reconciling the nutritional and glucocorticoid hypotheses of fetal programming. Faseb J 26(5):1866–1874PubMedCrossRef Cottrell EC, Holmes MC, Livingstone DE, Kenyon CJ, Seckl JR (2012) Reconciling the nutritional and glucocorticoid hypotheses of fetal programming. Faseb J 26(5):1866–1874PubMedCrossRef
48.
go back to reference Heussner K, Ruebner M, Huebner H, Rascher W, Menendez-Castro C, Hartner A, Fahlbusch FB, Rauh M (2015) Species differences of 11beta-hydroxysteroid dehydrogenase type 2 function in human and rat term placenta determined via LC-MS/MS. Placenta 37:79–84PubMedCrossRef Heussner K, Ruebner M, Huebner H, Rascher W, Menendez-Castro C, Hartner A, Fahlbusch FB, Rauh M (2015) Species differences of 11beta-hydroxysteroid dehydrogenase type 2 function in human and rat term placenta determined via LC-MS/MS. Placenta 37:79–84PubMedCrossRef
49.
go back to reference Lopez Bernal A, Craft IL (1981) Corticosteroid metabolism in vitro by human placenta, fetal membranes and decidua in early and late gestation. Placenta 2(4):279–285PubMedCrossRef Lopez Bernal A, Craft IL (1981) Corticosteroid metabolism in vitro by human placenta, fetal membranes and decidua in early and late gestation. Placenta 2(4):279–285PubMedCrossRef
51.
go back to reference Murphy BE (1981) Ontogeny of cortisol-cortisone interconversion in human tissues: a role for cortisone in human fetal development. J Steroid Biochem 14(9):811–817PubMedCrossRef Murphy BE (1981) Ontogeny of cortisol-cortisone interconversion in human tissues: a role for cortisone in human fetal development. J Steroid Biochem 14(9):811–817PubMedCrossRef
52.
go back to reference Murphy VE, Clifton VL (2003) Alterations in human placental 11beta-hydroxysteroid dehydrogenase type 1 and 2 with gestational age and labour. Placenta 24(7):739–744PubMedCrossRef Murphy VE, Clifton VL (2003) Alterations in human placental 11beta-hydroxysteroid dehydrogenase type 1 and 2 with gestational age and labour. Placenta 24(7):739–744PubMedCrossRef
53.
go back to reference Brown RW, Diaz R, Robson AC, Kotelevtsev YV, Mullins JJ, Kaufman MH, Seckl JR (1996) The ontogeny of 11 beta-hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development. Endocrinology 137(2):794–797PubMed Brown RW, Diaz R, Robson AC, Kotelevtsev YV, Mullins JJ, Kaufman MH, Seckl JR (1996) The ontogeny of 11 beta-hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development. Endocrinology 137(2):794–797PubMed
54.
go back to reference Clarke KA, Ward JW, Forhead AJ, Giussani DA, Fowden AL (2002) Regulation of 11 beta-hydroxysteroid dehydrogenase type 2 activity in ovine placenta by fetal cortisol. J Endocrinol 172(3):527–534PubMedCrossRef Clarke KA, Ward JW, Forhead AJ, Giussani DA, Fowden AL (2002) Regulation of 11 beta-hydroxysteroid dehydrogenase type 2 activity in ovine placenta by fetal cortisol. J Endocrinol 172(3):527–534PubMedCrossRef
55.
go back to reference Stewart PM, Murry BA, Mason JI (1994) Type 2 11 beta-hydroxysteroid dehydrogenase in human fetal tissues. J Clin Endocrinol Metab 78(6):1529–1532PubMed Stewart PM, Murry BA, Mason JI (1994) Type 2 11 beta-hydroxysteroid dehydrogenase in human fetal tissues. J Clin Endocrinol Metab 78(6):1529–1532PubMed
56.
go back to reference Pasqualini JR, Nguyen BL, Uhrich F, Wiqvist N, Diczfalusy E (1970) Cortisol and cortisone metabolism in the human foeto-placental unit at midgestation. J Steroid Biochem 1(1):209–219CrossRef Pasqualini JR, Nguyen BL, Uhrich F, Wiqvist N, Diczfalusy E (1970) Cortisol and cortisone metabolism in the human foeto-placental unit at midgestation. J Steroid Biochem 1(1):209–219CrossRef
57.
go back to reference Wyrwoll C, Keith M, Noble J, Stevenson PL, Bombail V, Crombie S, Evans LC, Bailey MA, Wood E, Seckl JR, Holmes MC (2015) Fetal brain 11beta-hydroxysteroid dehydrogenase type 2 selectively determines programming of adult depressive-like behaviors and cognitive function, but not anxiety behaviors in male mice. Psychoneuroendocrinology 59:59–70PubMedPubMedCentralCrossRef Wyrwoll C, Keith M, Noble J, Stevenson PL, Bombail V, Crombie S, Evans LC, Bailey MA, Wood E, Seckl JR, Holmes MC (2015) Fetal brain 11beta-hydroxysteroid dehydrogenase type 2 selectively determines programming of adult depressive-like behaviors and cognitive function, but not anxiety behaviors in male mice. Psychoneuroendocrinology 59:59–70PubMedPubMedCentralCrossRef
59.
go back to reference Kerzner LS, Stonestreet BS, Wu KY, Sadowska G, Malee MP (2002) Antenatal dexamethasone: effect on ovine placental 11beta-hydroxysteroid dehydrogenase type 2 expression and fetal growth. Pediatr Res 52(5):706–712. doi:10.1203/00006450-200211000-00016 PubMed Kerzner LS, Stonestreet BS, Wu KY, Sadowska G, Malee MP (2002) Antenatal dexamethasone: effect on ovine placental 11beta-hydroxysteroid dehydrogenase type 2 expression and fetal growth. Pediatr Res 52(5):706–712. doi:10.​1203/​00006450-200211000-00016 PubMed
61.
go back to reference Challis JR, Sloboda DM, Alfaidy N, Lye SJ, Gibb W, Patel FA, Whittle WL, Newnham JP (2002) Prostaglandins and mechanisms of preterm birth. Reproduction 124(1):1–17PubMedCrossRef Challis JR, Sloboda DM, Alfaidy N, Lye SJ, Gibb W, Patel FA, Whittle WL, Newnham JP (2002) Prostaglandins and mechanisms of preterm birth. Reproduction 124(1):1–17PubMedCrossRef
62.
go back to reference Chisaka H, Johnstone JF, Premyslova M, Manduch Z, Challis JR (2005) Effect of pro-inflammatory cytokines on expression and activity of 11beta-hydroxysteroid dehydrogenase type 2 in cultured human term placental trophoblast and human choriocarcinoma JEG-3 cells. J Soc Gynecol Investig 12(5):303–309. doi:10.1016/j.jsgi.2005.02.003 PubMedCrossRef Chisaka H, Johnstone JF, Premyslova M, Manduch Z, Challis JR (2005) Effect of pro-inflammatory cytokines on expression and activity of 11beta-hydroxysteroid dehydrogenase type 2 in cultured human term placental trophoblast and human choriocarcinoma JEG-3 cells. J Soc Gynecol Investig 12(5):303–309. doi:10.​1016/​j.​jsgi.​2005.​02.​003 PubMedCrossRef
63.
go back to reference Hardy DB, Yang K (2002) The expression of 11 beta-hydroxysteroid dehydrogenase type 2 is induced during trophoblast differentiation: effects of hypoxia. J Clin Endocrinol Metab 87(8):3696–3701. doi:10.1210/jcem.87.8.8720 PubMed Hardy DB, Yang K (2002) The expression of 11 beta-hydroxysteroid dehydrogenase type 2 is induced during trophoblast differentiation: effects of hypoxia. J Clin Endocrinol Metab 87(8):3696–3701. doi:10.​1210/​jcem.​87.​8.​8720 PubMed
64.
go back to reference Schoof E, Girstl M, Frobenius W, Kirschbaum M, Dorr HG, Rascher W, Dotsch J (2001) Decreased gene expression of 11beta-hydroxysteroid dehydrogenase type 2 and 15-hydroxyprostaglandin dehydrogenase in human placenta of patients with preeclampsia. J Clin Endocrinol Metab 86(3):1313–1317. doi:10.1210/jcem.86.3.7311 PubMed Schoof E, Girstl M, Frobenius W, Kirschbaum M, Dorr HG, Rascher W, Dotsch J (2001) Decreased gene expression of 11beta-hydroxysteroid dehydrogenase type 2 and 15-hydroxyprostaglandin dehydrogenase in human placenta of patients with preeclampsia. J Clin Endocrinol Metab 86(3):1313–1317. doi:10.​1210/​jcem.​86.​3.​7311 PubMed
65.
go back to reference Wyrwoll CS, Noble J, Thomson A, Tesic D, Miller MR, Rog-Zielinska EA, Moran CM, Seckl JR, Chapman KE, Holmes MC (2016) Pravastatin ameliorates placental vascular defects, fetal growth, and cardiac function in a model of glucocorticoid excess. Proc Natl Acad Sci U S A. doi:10.1073/pnas.1520356113 PubMed Wyrwoll CS, Noble J, Thomson A, Tesic D, Miller MR, Rog-Zielinska EA, Moran CM, Seckl JR, Chapman KE, Holmes MC (2016) Pravastatin ameliorates placental vascular defects, fetal growth, and cardiac function in a model of glucocorticoid excess. Proc Natl Acad Sci U S A. doi:10.​1073/​pnas.​1520356113 PubMed
66.
go back to reference Speirs HJ, Seckl JR, Brown RW (2004) Ontogeny of glucocorticoid receptor and 11beta-hydroxysteroid dehydrogenase type-1 gene expression identifies potential critical periods of glucocorticoid susceptibility during development. J Endocrinol 181(1):105–116PubMedCrossRef Speirs HJ, Seckl JR, Brown RW (2004) Ontogeny of glucocorticoid receptor and 11beta-hydroxysteroid dehydrogenase type-1 gene expression identifies potential critical periods of glucocorticoid susceptibility during development. J Endocrinol 181(1):105–116PubMedCrossRef
67.
go back to reference Waddell BJ, Benediktsson R, Brown RW, Seckl JR (1998) Tissue-specific messenger ribonucleic acid expression of 11beta-hydroxysteroid dehydrogenase types 1 and 2 and the glucocorticoid receptor within rat placenta suggests exquisite local control of glucocorticoid action. Endocrinology 139(4):1517–1523PubMed Waddell BJ, Benediktsson R, Brown RW, Seckl JR (1998) Tissue-specific messenger ribonucleic acid expression of 11beta-hydroxysteroid dehydrogenase types 1 and 2 and the glucocorticoid receptor within rat placenta suggests exquisite local control of glucocorticoid action. Endocrinology 139(4):1517–1523PubMed
68.
go back to reference Brown RW, Seckl JR (2005) Glucocorticoid action in development. Curr Opin Endocrinol Diabetes 12(3):224–232CrossRef Brown RW, Seckl JR (2005) Glucocorticoid action in development. Curr Opin Endocrinol Diabetes 12(3):224–232CrossRef
69.
go back to reference Hundertmark S, Dill A, Ebert A, Zimmermann B, Kotelevtsev YV, Mullins JJ, Seckl JR (2002) Foetal lung maturation in 11beta-hydroxysteroid dehydrogenase type 1 knockout mice. Horm Metab Res 34(10):545–549PubMedCrossRef Hundertmark S, Dill A, Ebert A, Zimmermann B, Kotelevtsev YV, Mullins JJ, Seckl JR (2002) Foetal lung maturation in 11beta-hydroxysteroid dehydrogenase type 1 knockout mice. Horm Metab Res 34(10):545–549PubMedCrossRef
70.
go back to reference Myatt L, Sun K (2010) Role of fetal membranes in signaling of fetal maturation and parturition. Int J Dev Biol 54(2–3):545–553PubMedCrossRef Myatt L, Sun K (2010) Role of fetal membranes in signaling of fetal maturation and parturition. Int J Dev Biol 54(2–3):545–553PubMedCrossRef
71.
go back to reference Yang Z, Guo C, Zhu P, Li W, Myatt L, Sun K (2007) Role of glucocorticoid receptor and CCAAT/enhancer-binding protein alpha in the feed-forward induction of 11beta-hydroxysteroid dehydrogenase type 1 expression by cortisol in human amnion fibroblasts. J Endocrinol 195(2):241–253PubMedCrossRef Yang Z, Guo C, Zhu P, Li W, Myatt L, Sun K (2007) Role of glucocorticoid receptor and CCAAT/enhancer-binding protein alpha in the feed-forward induction of 11beta-hydroxysteroid dehydrogenase type 1 expression by cortisol in human amnion fibroblasts. J Endocrinol 195(2):241–253PubMedCrossRef
72.
go back to reference Sun K, Myatt L (2003) Enhancement of glucocorticoid-induced 11beta-hydroxysteroid dehydrogenase type 1 expression by proinflammatory cytokines in cultured human amnion fibroblasts. Endocrinology 144(12):5568–5577PubMedCrossRef Sun K, Myatt L (2003) Enhancement of glucocorticoid-induced 11beta-hydroxysteroid dehydrogenase type 1 expression by proinflammatory cytokines in cultured human amnion fibroblasts. Endocrinology 144(12):5568–5577PubMedCrossRef
73.
go back to reference Reichardt HM, Schutz G (1996) Feedback control of glucocorticoid production is established during fetal development. Mol Med 2(6):735–744PubMedPubMedCentral Reichardt HM, Schutz G (1996) Feedback control of glucocorticoid production is established during fetal development. Mol Med 2(6):735–744PubMedPubMedCentral
74.
go back to reference Barlow SM, Morrison PJ, Sullivan FM (1974) Plasma corticosterone levels during pregnancy in the mouse: the relative contributions of the adrenal glands and foeto-placental units. J Endocrinol 60(3):473–483PubMedCrossRef Barlow SM, Morrison PJ, Sullivan FM (1974) Plasma corticosterone levels during pregnancy in the mouse: the relative contributions of the adrenal glands and foeto-placental units. J Endocrinol 60(3):473–483PubMedCrossRef
75.
go back to reference Hirasawa G, Takeyama J, Sasano H, Fukushima K, Suzuki T, Muramatu Y, Darnel AD, Kaneko C, Hiwatashi N, Toyota T, Nagura H, Krozowski ZS (2000) 11Beta-hydroxysteroid dehydrogenase type II and mineralocorticoid receptor in human placenta. J Clin Endocrinol Metab 85(3):1306–1309PubMed Hirasawa G, Takeyama J, Sasano H, Fukushima K, Suzuki T, Muramatu Y, Darnel AD, Kaneko C, Hiwatashi N, Toyota T, Nagura H, Krozowski ZS (2000) 11Beta-hydroxysteroid dehydrogenase type II and mineralocorticoid receptor in human placenta. J Clin Endocrinol Metab 85(3):1306–1309PubMed
76.
go back to reference Thompson A, Han VK, Yang K (2002) Spatial and temporal patterns of expression of 11beta-hydroxysteroid dehydrogenase types 1 and 2 messenger RNA and glucocorticoid receptor protein in the murine placenta and uterus during late pregnancy. Biol Reprod 67(6):1708–1718PubMedCrossRef Thompson A, Han VK, Yang K (2002) Spatial and temporal patterns of expression of 11beta-hydroxysteroid dehydrogenase types 1 and 2 messenger RNA and glucocorticoid receptor protein in the murine placenta and uterus during late pregnancy. Biol Reprod 67(6):1708–1718PubMedCrossRef
77.
go back to reference Rog-Zielinska EA, Thomson A, Kenyon CJ, Brownstein DG, Moran CM, Szumska D, Michailidou Z, Richardson J, Owen E, Watt A, Morrison H, Forrester LM, Bhattacharya S, Holmes MC, Chapman KE (2013) Glucocorticoid receptor is required for foetal heart maturation. Hum Mol Genet 22(16):3269–3282PubMedCrossRef Rog-Zielinska EA, Thomson A, Kenyon CJ, Brownstein DG, Moran CM, Szumska D, Michailidou Z, Richardson J, Owen E, Watt A, Morrison H, Forrester LM, Bhattacharya S, Holmes MC, Chapman KE (2013) Glucocorticoid receptor is required for foetal heart maturation. Hum Mol Genet 22(16):3269–3282PubMedCrossRef
78.
go back to reference Reul JM, Pearce PT, Funder JW, Krozowski ZS (1989) Type I and type II corticosteroid receptor gene expression in the rat: effect of adrenalectomy and dexamethasone administration. Mol Endocrinol 3(10):1674–1680. doi:10.1210/mend-3-10-1674 PubMedCrossRef Reul JM, Pearce PT, Funder JW, Krozowski ZS (1989) Type I and type II corticosteroid receptor gene expression in the rat: effect of adrenalectomy and dexamethasone administration. Mol Endocrinol 3(10):1674–1680. doi:10.​1210/​mend-3-10-1674 PubMedCrossRef
79.
go back to reference Ashwell JD, Lu FW, Vacchio MS (2000) Glucocorticoids in T cell development and function*. Annu Rev Immunol 18:309–345PubMedCrossRef Ashwell JD, Lu FW, Vacchio MS (2000) Glucocorticoids in T cell development and function*. Annu Rev Immunol 18:309–345PubMedCrossRef
80.
go back to reference Brewer JA, Sleckman BP, Swat W, Muglia LJ (2002) Green fluorescent protein-glucocorticoid receptor knockin mice reveal dynamic receptor modulation during thymocyte development. J Immunol 169(3):1309–1318PubMedCrossRef Brewer JA, Sleckman BP, Swat W, Muglia LJ (2002) Green fluorescent protein-glucocorticoid receptor knockin mice reveal dynamic receptor modulation during thymocyte development. J Immunol 169(3):1309–1318PubMedCrossRef
81.
go back to reference Holleman A, Cheok MH, den Boer ML, Yang W, Veerman AJ, Kazemier KM, Pei D, Cheng C, Pui CH, Relling MV, Janka-Schaub GE, Pieters R, Evans WE (2004) Gene-expression patterns in drug-resistant acute lymphoblastic leukemia cells and response to treatment. N Engl J Med 351(6):533–542PubMedCrossRef Holleman A, Cheok MH, den Boer ML, Yang W, Veerman AJ, Kazemier KM, Pei D, Cheng C, Pui CH, Relling MV, Janka-Schaub GE, Pieters R, Evans WE (2004) Gene-expression patterns in drug-resistant acute lymphoblastic leukemia cells and response to treatment. N Engl J Med 351(6):533–542PubMedCrossRef
82.
go back to reference Hulleman E, Kazemier KM, Holleman A, VanderWeele DJ, Rudin CM, Broekhuis MJ, Evans WE, Pieters R, Den Boer ML (2009) Inhibition of glycolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood 113(9):2014–2021PubMedPubMedCentralCrossRef Hulleman E, Kazemier KM, Holleman A, VanderWeele DJ, Rudin CM, Broekhuis MJ, Evans WE, Pieters R, Den Boer ML (2009) Inhibition of glycolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood 113(9):2014–2021PubMedPubMedCentralCrossRef
83.
go back to reference Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Passier R, Mummery CL (2015) Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function. Cell Rep 13(4):733–745PubMedPubMedCentralCrossRef Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Passier R, Mummery CL (2015) Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function. Cell Rep 13(4):733–745PubMedPubMedCentralCrossRef
84.
go back to reference Rog-Zielinska EA, Craig MA, Manning JR, Richardson RV, Gowans GJ, Dunbar DR, Gharbi K, Kenyon CJ, Holmes MC, Hardie DG, Smith GL, Chapman KE (2014) Glucocorticoids promote structural and functional maturation of foetal cardiomyocytes: a role for PGC-1alpha. Cell Death Differ 22(7):1106–1116PubMedPubMedCentralCrossRef Rog-Zielinska EA, Craig MA, Manning JR, Richardson RV, Gowans GJ, Dunbar DR, Gharbi K, Kenyon CJ, Holmes MC, Hardie DG, Smith GL, Chapman KE (2014) Glucocorticoids promote structural and functional maturation of foetal cardiomyocytes: a role for PGC-1alpha. Cell Death Differ 22(7):1106–1116PubMedPubMedCentralCrossRef
85.
go back to reference Thomassin H, Flavin M, Espinas ML, Grange T (2001) Glucocorticoid-induced DNA demethylation and gene memory during development. EMBO J 20(8):1974–1983PubMedPubMedCentralCrossRef Thomassin H, Flavin M, Espinas ML, Grange T (2001) Glucocorticoid-induced DNA demethylation and gene memory during development. EMBO J 20(8):1974–1983PubMedPubMedCentralCrossRef
86.
go back to reference Seckl JR, Holmes MC (2007) Mechanisms of disease: glucocorticoids, their placental metabolism and fetal ‘programming’ of adult pathophysiology. Nat Clin Pract Endocrinol Metab 3(6):479–488. doi:10.1038/ncpendmet0515 PubMedCrossRef Seckl JR, Holmes MC (2007) Mechanisms of disease: glucocorticoids, their placental metabolism and fetal ‘programming’ of adult pathophysiology. Nat Clin Pract Endocrinol Metab 3(6):479–488. doi:10.​1038/​ncpendmet0515 PubMedCrossRef
87.
go back to reference Weinstock M (2008) The long-term behavioural consequences of prenatal stress. Neurosci Biobehav Rev 32:1073–1086PubMedCrossRef Weinstock M (2008) The long-term behavioural consequences of prenatal stress. Neurosci Biobehav Rev 32:1073–1086PubMedCrossRef
88.
go back to reference Vallee M, Mayo W, Dellu F, LeMoal M, Simon H, Maccari S (1997) Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion. J Neurosci 17:2626–2636PubMed Vallee M, Mayo W, Dellu F, LeMoal M, Simon H, Maccari S (1997) Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion. J Neurosci 17:2626–2636PubMed
89.
go back to reference Henry C, Kabbaj M, Simon H, Le Moal M, Maccari S (1994) Prenatal stress increases the hypothalamo-pituitary-adrenal axis response in young and adult rats. J Neuroendocrinol 6(3):341–345PubMedCrossRef Henry C, Kabbaj M, Simon H, Le Moal M, Maccari S (1994) Prenatal stress increases the hypothalamo-pituitary-adrenal axis response in young and adult rats. J Neuroendocrinol 6(3):341–345PubMedCrossRef
90.
go back to reference Pincus-Knackstedt MK, Joachim RA, Blois SM, Douglas AJ, Orsal AS, Klapp BF, Wahn U, Hamelmann E, Arck PC (2006) Prenatal stress enhances susceptibility of murine adult offspring toward airway inflammation. J Immunol 177(12):8484–8492PubMedCrossRef Pincus-Knackstedt MK, Joachim RA, Blois SM, Douglas AJ, Orsal AS, Klapp BF, Wahn U, Hamelmann E, Arck PC (2006) Prenatal stress enhances susceptibility of murine adult offspring toward airway inflammation. J Immunol 177(12):8484–8492PubMedCrossRef
92.
93.
go back to reference Louey S, Thornburg KL (2005) The prenatal environment and later cardiovascular disease. Early Hum Dev 81:745–751PubMedCrossRef Louey S, Thornburg KL (2005) The prenatal environment and later cardiovascular disease. Early Hum Dev 81:745–751PubMedCrossRef
94.
go back to reference Moisiadis VG, Matthews SG (2014) Glucocorticoids and fetal programming part 1: outcomes. Nat Rev Endocrinol 10(7):391–402PubMedCrossRef Moisiadis VG, Matthews SG (2014) Glucocorticoids and fetal programming part 1: outcomes. Nat Rev Endocrinol 10(7):391–402PubMedCrossRef
95.
go back to reference White PC, Agarwal AK, Nunez BS, Giacchetti G, Mantero F, Stewart PM (2000) Genotype-phenotype correlations of mutations and polymorphisms in HSD11B2, the gene encoding the kidney isozyme of 11beta-hydroxysteroid dehydrogenase. Endocr Res 26:771–780PubMedCrossRef White PC, Agarwal AK, Nunez BS, Giacchetti G, Mantero F, Stewart PM (2000) Genotype-phenotype correlations of mutations and polymorphisms in HSD11B2, the gene encoding the kidney isozyme of 11beta-hydroxysteroid dehydrogenase. Endocr Res 26:771–780PubMedCrossRef
96.
go back to reference Solano ME, Kowal MK, O’Rourke GE, Horst AK, Modest K, Plosch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC (2015) Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 125(4):1726–1738. doi:10.1172/JCI68140 PubMedPubMedCentralCrossRef Solano ME, Kowal MK, O’Rourke GE, Horst AK, Modest K, Plosch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC (2015) Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 125(4):1726–1738. doi:10.​1172/​JCI68140 PubMedPubMedCentralCrossRef
97.
go back to reference Nugent JL, Wareing M, Palin V, Sibley CP, Baker PN, Ray DW, Farrow SN, Jones RL (2013) Chronic glucocorticoid exposure potentiates placental chorionic plate artery constriction: implications for aberrant fetoplacental vascular resistance in fetal growth restriction. Endocrinology 154(2):876–887PubMedCrossRef Nugent JL, Wareing M, Palin V, Sibley CP, Baker PN, Ray DW, Farrow SN, Jones RL (2013) Chronic glucocorticoid exposure potentiates placental chorionic plate artery constriction: implications for aberrant fetoplacental vascular resistance in fetal growth restriction. Endocrinology 154(2):876–887PubMedCrossRef
98.
go back to reference Nyirenda MJ, Lindsay RS, Kenyon CJ, Burchell A, Seckl JR (1998) Glucocorticoid exposure in late gestation permanently programs rat hepatic phosphoenolpyruvate carboxykinase and glucocorticoid receptor expression and causes glucose intolerance in adult offspring. J Clin Invest 101(10):2174–2181PubMedPubMedCentralCrossRef Nyirenda MJ, Lindsay RS, Kenyon CJ, Burchell A, Seckl JR (1998) Glucocorticoid exposure in late gestation permanently programs rat hepatic phosphoenolpyruvate carboxykinase and glucocorticoid receptor expression and causes glucose intolerance in adult offspring. J Clin Invest 101(10):2174–2181PubMedPubMedCentralCrossRef
99.
go back to reference Yehuda R, Engel SM, Brand SR, Seckl J, Marcus SM, Berkowitz G (2005) Transgenerational effects of posttraumatic stress disorder in babies of mothers exposed to the World Trade Center attacks during pregnancy. J Clin Endocrinol Metab 90:4115–4118PubMedCrossRef Yehuda R, Engel SM, Brand SR, Seckl J, Marcus SM, Berkowitz G (2005) Transgenerational effects of posttraumatic stress disorder in babies of mothers exposed to the World Trade Center attacks during pregnancy. J Clin Endocrinol Metab 90:4115–4118PubMedCrossRef
100.
go back to reference Cole TJ, Blendy JA, Monaghan AP, Krieglstein K, Schmid W, Aguzzi A, Fantuzzi G, Hummler E, Unsicker K, Schutz G (1995) Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation. Genes Dev 9(13):1608–1621PubMedCrossRef Cole TJ, Blendy JA, Monaghan AP, Krieglstein K, Schmid W, Aguzzi A, Fantuzzi G, Hummler E, Unsicker K, Schutz G (1995) Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation. Genes Dev 9(13):1608–1621PubMedCrossRef
101.
go back to reference Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, Lightman SL (2000) Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proc Natl Acad Sci U S A 97:5645–5650PubMedPubMedCentralCrossRef Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, Lightman SL (2000) Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proc Natl Acad Sci U S A 97:5645–5650PubMedPubMedCentralCrossRef
103.
go back to reference Wyrwoll CS, Holmes MC, Seckl JR (2011) 11b-Hydroxysteroid dehydrogenases and the brain: from zero to hero, a decade of progress. Front Neuroendocrinol 32:265–286PubMedPubMedCentralCrossRef Wyrwoll CS, Holmes MC, Seckl JR (2011) 11b-Hydroxysteroid dehydrogenases and the brain: from zero to hero, a decade of progress. Front Neuroendocrinol 32:265–286PubMedPubMedCentralCrossRef
104.
go back to reference Coutinho AE, Chapman KE (2011) The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol Cell Endocrinol 335(1):2–13PubMedPubMedCentralCrossRef Coutinho AE, Chapman KE (2011) The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol Cell Endocrinol 335(1):2–13PubMedPubMedCentralCrossRef
105.
go back to reference McEwen BS, Biron CA, Brunson KW, Bulloch K, Chambers WH, Dhabhar FS, Goldfarb RH, Kitson RP, Miller AH, Spencer RL, Weiss JM (1997) The role of adrenocorticoids as modulators of immune function in health and disease: neural, endocrine and immune interactions. Brain Res Brain Res Rev 23(1–2):79–133PubMedCrossRef McEwen BS, Biron CA, Brunson KW, Bulloch K, Chambers WH, Dhabhar FS, Goldfarb RH, Kitson RP, Miller AH, Spencer RL, Weiss JM (1997) The role of adrenocorticoids as modulators of immune function in health and disease: neural, endocrine and immune interactions. Brain Res Brain Res Rev 23(1–2):79–133PubMedCrossRef
106.
go back to reference Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, Fischer D, Holsboer F, Wotjak CT, Almeida OF, Spengler D (2009) Dynamic DNA methylation programs persistent adverse effects of early-life stress. Nat Neurosci 12(12):1559–1566PubMedCrossRef Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, Fischer D, Holsboer F, Wotjak CT, Almeida OF, Spengler D (2009) Dynamic DNA methylation programs persistent adverse effects of early-life stress. Nat Neurosci 12(12):1559–1566PubMedCrossRef
107.
go back to reference Brydges NM, Jin R, Seckl J, Holmes MC, Drake AJ, Hall J (2014) Juvenile stress enhances anxiety and alters corticosteroid receptor expression in adulthood. Brain Behav 4(1):4–13PubMedCrossRef Brydges NM, Jin R, Seckl J, Holmes MC, Drake AJ, Hall J (2014) Juvenile stress enhances anxiety and alters corticosteroid receptor expression in adulthood. Brain Behav 4(1):4–13PubMedCrossRef
108.
go back to reference Cai G, Ziko I, Barwood J, Soch A, Sominsky L, Molero JC, Spencer SJ (2016) Overfeeding during a critical postnatal period exacerbates hypothalamic-pituitary-adrenal axis responses to immune challenge: a role for adrenal melanocortin 2 receptors. Sci Rep 6:21097PubMedPubMedCentralCrossRef Cai G, Ziko I, Barwood J, Soch A, Sominsky L, Molero JC, Spencer SJ (2016) Overfeeding during a critical postnatal period exacerbates hypothalamic-pituitary-adrenal axis responses to immune challenge: a role for adrenal melanocortin 2 receptors. Sci Rep 6:21097PubMedPubMedCentralCrossRef
109.
go back to reference Fuenfer MM, Herson VC, Raye JR, Woronick CL, Eisenfeld L, Ingardia CJ, Block CF, Krause PJ (1987) The effect of betamethasone on neonatal neutrophil chemotaxis. Pediatr Res 22(2):150–153PubMedCrossRef Fuenfer MM, Herson VC, Raye JR, Woronick CL, Eisenfeld L, Ingardia CJ, Block CF, Krause PJ (1987) The effect of betamethasone on neonatal neutrophil chemotaxis. Pediatr Res 22(2):150–153PubMedCrossRef
110.
go back to reference Barak M, Cohen A, Herschkowitz S (1992) Total leukocyte and neutrophil count changes associated with antenatal betamethasone administration in premature infants. Acta Paediatr 81(10):760–763PubMedCrossRef Barak M, Cohen A, Herschkowitz S (1992) Total leukocyte and neutrophil count changes associated with antenatal betamethasone administration in premature infants. Acta Paediatr 81(10):760–763PubMedCrossRef
111.
go back to reference Caldas JP, Vilela MM, Braghini CA, Mazzola TN, Marba ST (2012) Antenatal maternal corticosteroid administration and markers of oxidative stress and inflammation in umbilical cord blood from very low birth weight preterm newborn infants. J Pediatr (Rio J) 88(1):61–66CrossRef Caldas JP, Vilela MM, Braghini CA, Mazzola TN, Marba ST (2012) Antenatal maternal corticosteroid administration and markers of oxidative stress and inflammation in umbilical cord blood from very low birth weight preterm newborn infants. J Pediatr (Rio J) 88(1):61–66CrossRef
113.
go back to reference Kavelaars A, van der Pompe G, Bakker JM, van Hasselt PM, Cats B, Visser GH, Heijnen CJ (1999) Altered immune function in human newborns after prenatal administration of betamethasone: enhanced natural killer cell activity and decreased T cell proliferation in cord blood. Pediatr Res 45(3):306–312PubMedCrossRef Kavelaars A, van der Pompe G, Bakker JM, van Hasselt PM, Cats B, Visser GH, Heijnen CJ (1999) Altered immune function in human newborns after prenatal administration of betamethasone: enhanced natural killer cell activity and decreased T cell proliferation in cord blood. Pediatr Res 45(3):306–312PubMedCrossRef
114.
go back to reference Chabra S, Cottrill C, Rayens MK, Cross R, Lipke D, Bruce M (1998) Lymphocyte subsets in cord blood of preterm infants: effect of antenatal steroids. Biol Neonate 74(3):200–207PubMedCrossRef Chabra S, Cottrill C, Rayens MK, Cross R, Lipke D, Bruce M (1998) Lymphocyte subsets in cord blood of preterm infants: effect of antenatal steroids. Biol Neonate 74(3):200–207PubMedCrossRef
115.
go back to reference Agakidis C, Sarafidis K, Tzimouli V, Agakidou E, Taparkou A, Kanakoudi-Tsakalidou F, Soubasi-Griva V (2009) Antenatal betamethasone does not influence lymphocyte apoptosis in preterm neonates. Am J Perinatol 26(7):485–490PubMedCrossRef Agakidis C, Sarafidis K, Tzimouli V, Agakidou E, Taparkou A, Kanakoudi-Tsakalidou F, Soubasi-Griva V (2009) Antenatal betamethasone does not influence lymphocyte apoptosis in preterm neonates. Am J Perinatol 26(7):485–490PubMedCrossRef
118.
go back to reference Slack MH, Schapira D, Thwaites RJ, Schapira C, Bamber J, Burrage M, Southern J, Andrews N, Miller E (2004) Acellular pertussis vaccine given by accelerated schedule: response of preterm infants. Arch Dis Child Fetal Neonatal Ed 89(1):F57–60PubMedPubMedCentralCrossRef Slack MH, Schapira D, Thwaites RJ, Schapira C, Bamber J, Burrage M, Southern J, Andrews N, Miller E (2004) Acellular pertussis vaccine given by accelerated schedule: response of preterm infants. Arch Dis Child Fetal Neonatal Ed 89(1):F57–60PubMedPubMedCentralCrossRef
120.
go back to reference Wright RJ, Visness CM, Calatroni A, Grayson MH, Gold DR, Sandel MT, Lee-Parritz A, Wood RA, Kattan M, Bloomberg GR, Burger M, Togias A, Witter FR, Sperling RS, Sadovsky Y, Gern JE (2010) Prenatal maternal stress and cord blood innate and adaptive cytokine responses in an inner-city cohort. Am J Respir Crit Care Med 182(1):25–33. doi:10.1164/rccm.200904-0637OC PubMedPubMedCentralCrossRef Wright RJ, Visness CM, Calatroni A, Grayson MH, Gold DR, Sandel MT, Lee-Parritz A, Wood RA, Kattan M, Bloomberg GR, Burger M, Togias A, Witter FR, Sperling RS, Sadovsky Y, Gern JE (2010) Prenatal maternal stress and cord blood innate and adaptive cytokine responses in an inner-city cohort. Am J Respir Crit Care Med 182(1):25–33. doi:10.​1164/​rccm.​200904-0637OC PubMedPubMedCentralCrossRef
124.
go back to reference Wen HJ, Wang YJ, Lin YC, Chang CC, Shieh CC, Lung FW, Guo YL (2011) Prediction of atopic dermatitis in 2-yr-old children by cord blood IgE, genetic polymorphisms in cytokine genes, and maternal mentality during pregnancy. Pediatr Allergy Immunol 22(7):695–703. doi:10.1111/j.1399-3038.2011.01177.x PubMedCrossRef Wen HJ, Wang YJ, Lin YC, Chang CC, Shieh CC, Lung FW, Guo YL (2011) Prediction of atopic dermatitis in 2-yr-old children by cord blood IgE, genetic polymorphisms in cytokine genes, and maternal mentality during pregnancy. Pediatr Allergy Immunol 22(7):695–703. doi:10.​1111/​j.​1399-3038.​2011.​01177.​x PubMedCrossRef
126.
go back to reference Chiu YH, Coull BA, Sternthal MJ, Kloog I, Schwartz J, Cohen S, Wright RJ (2014) Effects of prenatal community violence and ambient air pollution on childhood wheeze in an urban population. J Allergy Clin Immunol 133(3):713–722 e714. doi:10.1016/j.jaci.2013.09.023 PubMedCrossRef Chiu YH, Coull BA, Sternthal MJ, Kloog I, Schwartz J, Cohen S, Wright RJ (2014) Effects of prenatal community violence and ambient air pollution on childhood wheeze in an urban population. J Allergy Clin Immunol 133(3):713–722 e714. doi:10.​1016/​j.​jaci.​2013.​09.​023 PubMedCrossRef
128.
go back to reference Guxens M, Sonnenschein-van der Voort AM, Tiemeier H, Hofman A, Sunyer J, de Jongste JC, Jaddoe VW, Duijts L (2014) Parental psychological distress during pregnancy and wheezing in preschool children: the Generation R Study. J Allergy Clin Immunol 133(1):59–67 e51-12. doi:10.1016/j.jaci.2013.04.044 PubMedCrossRef Guxens M, Sonnenschein-van der Voort AM, Tiemeier H, Hofman A, Sunyer J, de Jongste JC, Jaddoe VW, Duijts L (2014) Parental psychological distress during pregnancy and wheezing in preschool children: the Generation R Study. J Allergy Clin Immunol 133(1):59–67 e51-12. doi:10.​1016/​j.​jaci.​2013.​04.​044 PubMedCrossRef
129.
go back to reference Lee A, Mathilda Chiu YH, Rosa MJ, Jara C, Wright RO, Coull BA, Wright RJ (2016) Prenatal and postnatal stress and asthma in children: temporal- and sex-specific associations. J Allergy Clin Immunol. doi:10.1016/j.jaci.2016.01.014 Lee A, Mathilda Chiu YH, Rosa MJ, Jara C, Wright RO, Coull BA, Wright RJ (2016) Prenatal and postnatal stress and asthma in children: temporal- and sex-specific associations. J Allergy Clin Immunol. doi:10.​1016/​j.​jaci.​2016.​01.​014
130.
go back to reference Larsen AD, Schlunssen V, Christensen BH, Bonde JP, Obel C, Thulstrup AM, Hannerz H, Hougaard KS (2014) Exposure to psychosocial job strain during pregnancy and odds of asthma and atopic dermatitis among 7-year old children—a prospective cohort study. Scand J Work Environ Health 40(6):639–648. doi:10.5271/sjweh.3452 PubMedCrossRef Larsen AD, Schlunssen V, Christensen BH, Bonde JP, Obel C, Thulstrup AM, Hannerz H, Hougaard KS (2014) Exposure to psychosocial job strain during pregnancy and odds of asthma and atopic dermatitis among 7-year old children—a prospective cohort study. Scand J Work Environ Health 40(6):639–648. doi:10.​5271/​sjweh.​3452 PubMedCrossRef
131.
go back to reference Pole JD, Mustard CA, To T, Beyene J, Allen AC (2010) Antenatal steroid therapy for fetal lung maturation and the subsequent risk of childhood asthma: a longitudinal analysis. J Pregnancy 2010:789748PubMedPubMedCentralCrossRef Pole JD, Mustard CA, To T, Beyene J, Allen AC (2010) Antenatal steroid therapy for fetal lung maturation and the subsequent risk of childhood asthma: a longitudinal analysis. J Pregnancy 2010:789748PubMedPubMedCentralCrossRef
132.
134.
go back to reference Hartwig IR, Bruenahl CA, Ramisch K, Keil T, Inman M, Arck PC, Pincus M (2014) Reduced levels of maternal progesterone during pregnancy increase the risk for allergic airway diseases in females only. J Mol Med (Berl) 92(10):1093–1104. doi:10.1007/s00109-014-1167-9 CrossRef Hartwig IR, Bruenahl CA, Ramisch K, Keil T, Inman M, Arck PC, Pincus M (2014) Reduced levels of maternal progesterone during pregnancy increase the risk for allergic airway diseases in females only. J Mol Med (Berl) 92(10):1093–1104. doi:10.​1007/​s00109-014-1167-9 CrossRef
136.
go back to reference Vermillion ST, Soper DE, Newman RB (2000) Neonatal sepsis and death after multiple courses of antenatal betamethasone therapy. Am J Obstet Gynecol 183(4):810–814PubMedCrossRef Vermillion ST, Soper DE, Newman RB (2000) Neonatal sepsis and death after multiple courses of antenatal betamethasone therapy. Am J Obstet Gynecol 183(4):810–814PubMedCrossRef
138.
go back to reference Smolders-de Haas H, Neuvel J, Schmand B, Treffers PE, Koppe JG, Hoeks J (1990) Physical development and medical history of children who were treated antenatally with corticosteroids to prevent respiratory distress syndrome: a 10- to 12-year follow-up. Pediatrics 86(1):65–70PubMed Smolders-de Haas H, Neuvel J, Schmand B, Treffers PE, Koppe JG, Hoeks J (1990) Physical development and medical history of children who were treated antenatally with corticosteroids to prevent respiratory distress syndrome: a 10- to 12-year follow-up. Pediatrics 86(1):65–70PubMed
140.
go back to reference Greene NH, Pedersen LH, Liu S, Olsen J (2013) Prenatal prescription corticosteroids and offspring diabetes: a national cohort study. Int J Epidemiol 42(1):186–193PubMedCrossRef Greene NH, Pedersen LH, Liu S, Olsen J (2013) Prenatal prescription corticosteroids and offspring diabetes: a national cohort study. Int J Epidemiol 42(1):186–193PubMedCrossRef
142.
go back to reference Li P, Tong Y, Yang H, Zhou S, Xiong F, Huo T, Mao M (2014) Mitochondrial translocation of human telomerase reverse transcriptase in cord blood mononuclear cells of newborns with gestational diabetes mellitus mothers. Diabetes Res Clin Pract 103(2):310–318PubMedCrossRef Li P, Tong Y, Yang H, Zhou S, Xiong F, Huo T, Mao M (2014) Mitochondrial translocation of human telomerase reverse transcriptase in cord blood mononuclear cells of newborns with gestational diabetes mellitus mothers. Diabetes Res Clin Pract 103(2):310–318PubMedCrossRef
146.
149.
go back to reference Liang L, Willis-Owen SA, Laprise C, Wong KC, Davies GA, Hudson TJ, Binia A, Hopkin JM, Yang IV, Grundberg E, Busche S, Hudson M, Ronnblom L, Pastinen TM, Schwartz DA, Lathrop GM, Moffatt MF, Cookson WO (2015) An epigenome-wide association study of total serum immunoglobulin E concentration. Nature 520(7549):670–674. doi:10.1038/nature14125 PubMedPubMedCentralCrossRef Liang L, Willis-Owen SA, Laprise C, Wong KC, Davies GA, Hudson TJ, Binia A, Hopkin JM, Yang IV, Grundberg E, Busche S, Hudson M, Ronnblom L, Pastinen TM, Schwartz DA, Lathrop GM, Moffatt MF, Cookson WO (2015) An epigenome-wide association study of total serum immunoglobulin E concentration. Nature 520(7549):670–674. doi:10.​1038/​nature14125 PubMedPubMedCentralCrossRef
150.
go back to reference Tolosa E, Ashwell JD (1999) Thymus-derived glucocorticoids and the regulation of antigen-specific T-cell development. Neuroimmunomodulation 6(1–2):90–96PubMedCrossRef Tolosa E, Ashwell JD (1999) Thymus-derived glucocorticoids and the regulation of antigen-specific T-cell development. Neuroimmunomodulation 6(1–2):90–96PubMedCrossRef
151.
go back to reference Breant B, Gesina E, Blondeau B (2006) Nutrition, glucocorticoids and pancreas development. Horm Res 65(Suppl 3):98–104PubMedCrossRef Breant B, Gesina E, Blondeau B (2006) Nutrition, glucocorticoids and pancreas development. Horm Res 65(Suppl 3):98–104PubMedCrossRef
152.
go back to reference Santner-Nanan B, Straubinger K, Hsu P, Parnell G, Tang B, Xu B, Makris A, Hennessy A, Peek MJ, Busch DH, da Costa CP, Nanan R (2013) Fetal-maternal alignment of regulatory T cells correlates with IL-10 and Bcl-2 upregulation in pregnancy. J Immunol 191(1):145–153. doi:10.4049/jimmunol.1203165 PubMedPubMedCentralCrossRef Santner-Nanan B, Straubinger K, Hsu P, Parnell G, Tang B, Xu B, Makris A, Hennessy A, Peek MJ, Busch DH, da Costa CP, Nanan R (2013) Fetal-maternal alignment of regulatory T cells correlates with IL-10 and Bcl-2 upregulation in pregnancy. J Immunol 191(1):145–153. doi:10.​4049/​jimmunol.​1203165 PubMedPubMedCentralCrossRef
153.
go back to reference Malhotra I, Ouma J, Wamachi A, Kioko J, Mungai P, Omollo A, Elson L, Koech D, Kazura JW, King CL (1997) In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest 99(7):1759–1766PubMedPubMedCentralCrossRef Malhotra I, Ouma J, Wamachi A, Kioko J, Mungai P, Omollo A, Elson L, Koech D, Kazura JW, King CL (1997) In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest 99(7):1759–1766PubMedPubMedCentralCrossRef
156.
go back to reference Coskun S, Chao H, Vasavada H, Heydari K, Gonzales N, Zhou X, de Crombrugghe B, Hirschi KK (2014) Development of the fetal bone marrow niche and regulation of HSC quiescence and homing ability by emerging osteolineage cells. Cell Rep 9(2):581–590PubMedPubMedCentralCrossRef Coskun S, Chao H, Vasavada H, Heydari K, Gonzales N, Zhou X, de Crombrugghe B, Hirschi KK (2014) Development of the fetal bone marrow niche and regulation of HSC quiescence and homing ability by emerging osteolineage cells. Cell Rep 9(2):581–590PubMedPubMedCentralCrossRef
157.
go back to reference Mikkola HK, Orkin SH (2006) The journey of developing hematopoietic stem cells. Development 133(19):3733–3744PubMedCrossRef Mikkola HK, Orkin SH (2006) The journey of developing hematopoietic stem cells. Development 133(19):3733–3744PubMedCrossRef
158.
go back to reference Beerman I, Bock C, Garrison BS, Smith ZD, Gu H, Meissner A, Rossi DJ (2013) Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12(4):413–425PubMedCrossRef Beerman I, Bock C, Garrison BS, Smith ZD, Gu H, Meissner A, Rossi DJ (2013) Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12(4):413–425PubMedCrossRef
159.
go back to reference Igarashi H, Kouro T, Yokota T, Comp PC, Kincade PW (2001) Age and stage dependency of estrogen receptor expression by lymphocyte precursors. Proc Natl Acad Sci U S A 98(26):15131–15136PubMedPubMedCentralCrossRef Igarashi H, Kouro T, Yokota T, Comp PC, Kincade PW (2001) Age and stage dependency of estrogen receptor expression by lymphocyte precursors. Proc Natl Acad Sci U S A 98(26):15131–15136PubMedPubMedCentralCrossRef
160.
go back to reference Kollet O, Vagima Y, D’Uva G, Golan K, Canaani J, Itkin T, Gur-Cohen S, Kalinkovich A, Caglio G, Medaglia C, Ludin A, Lapid K, Shezen E, Neufeld-Cohen A, Varol D, Chen A, Lapidot T (2013) Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors. Leukemia 27(10):2006–2015PubMedCrossRef Kollet O, Vagima Y, D’Uva G, Golan K, Canaani J, Itkin T, Gur-Cohen S, Kalinkovich A, Caglio G, Medaglia C, Ludin A, Lapid K, Shezen E, Neufeld-Cohen A, Varol D, Chen A, Lapidot T (2013) Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors. Leukemia 27(10):2006–2015PubMedCrossRef
161.
go back to reference Gekas C, Dieterlen-Lievre F, Orkin SH, Mikkola HK (2005) The placenta is a niche for hematopoietic stem cells. Dev Cell 8(3):365–375PubMedCrossRef Gekas C, Dieterlen-Lievre F, Orkin SH, Mikkola HK (2005) The placenta is a niche for hematopoietic stem cells. Dev Cell 8(3):365–375PubMedCrossRef
162.
go back to reference Medvinsky A, Rybtsov S, Taoudi S (2011) Embryonic origin of the adult hematopoietic system: advances and questions. Development 138(6):1017–1031PubMedCrossRef Medvinsky A, Rybtsov S, Taoudi S (2011) Embryonic origin of the adult hematopoietic system: advances and questions. Development 138(6):1017–1031PubMedCrossRef
163.
go back to reference Heinig K, Sage F, Robin C, Sperandio M (2015) Development and trafficking function of haematopoietic stem cells and myeloid cells during fetal ontogeny. Cardiovasc Res 107(3):352–363PubMedCrossRef Heinig K, Sage F, Robin C, Sperandio M (2015) Development and trafficking function of haematopoietic stem cells and myeloid cells during fetal ontogeny. Cardiovasc Res 107(3):352–363PubMedCrossRef
164.
go back to reference Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald HR (2015) Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518(7540):547–551PubMedCrossRef Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald HR (2015) Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518(7540):547–551PubMedCrossRef
165.
go back to reference Rhodes KE, Gekas C, Wang Y, Lux CT, Francis CS, Chan DN, Conway S, Orkin SH, Yoder MC, Mikkola HK (2008) The emergence of hematopoietic stem cells is initiated in the placental vasculature in the absence of circulation. Cell Stem Cell 2(3):252–263PubMedPubMedCentralCrossRef Rhodes KE, Gekas C, Wang Y, Lux CT, Francis CS, Chan DN, Conway S, Orkin SH, Yoder MC, Mikkola HK (2008) The emergence of hematopoietic stem cells is initiated in the placental vasculature in the absence of circulation. Cell Stem Cell 2(3):252–263PubMedPubMedCentralCrossRef
166.
go back to reference Kumaravelu P, Hook L, Morrison AM, Ure J, Zhao S, Zuyev S, Ansell J, Medvinsky A (2002) Quantitative developmental anatomy of definitive haematopoietic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver. Development 129(21):4891–4899PubMed Kumaravelu P, Hook L, Morrison AM, Ure J, Zhao S, Zuyev S, Ansell J, Medvinsky A (2002) Quantitative developmental anatomy of definitive haematopoietic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver. Development 129(21):4891–4899PubMed
168.
169.
go back to reference Khan JA, Mendelson A, Kunisaki Y, Birbrair A, Kou Y, Arnal-Estape A, Pinho S, Ciero P, Nakahara F, Ma’ayan A, Bergman A, Merad M, Frenette PS (2016) Fetal liver hematopoietic stem cell niches associate with portal vessels. Science 351(6269):176–180PubMedCrossRef Khan JA, Mendelson A, Kunisaki Y, Birbrair A, Kou Y, Arnal-Estape A, Pinho S, Ciero P, Nakahara F, Ma’ayan A, Bergman A, Merad M, Frenette PS (2016) Fetal liver hematopoietic stem cell niches associate with portal vessels. Science 351(6269):176–180PubMedCrossRef
170.
go back to reference Ema H, Nakauchi H (2000) Expansion of hematopoietic stem cells in the developing liver of a mouse embryo. Blood 95(7):2284–2288PubMed Ema H, Nakauchi H (2000) Expansion of hematopoietic stem cells in the developing liver of a mouse embryo. Blood 95(7):2284–2288PubMed
171.
go back to reference Kinoshita T, Sekiguchi T, Xu MJ, Ito Y, Kamiya A, Tsuji K, Nakahata T, Miyajima A (1999) Hepatic differentiation induced by oncostatin M attenuates fetal liver hematopoiesis. Proc Natl Acad Sci U S A 96(13):7265–7270PubMedPubMedCentralCrossRef Kinoshita T, Sekiguchi T, Xu MJ, Ito Y, Kamiya A, Tsuji K, Nakahata T, Miyajima A (1999) Hepatic differentiation induced by oncostatin M attenuates fetal liver hematopoiesis. Proc Natl Acad Sci U S A 96(13):7265–7270PubMedPubMedCentralCrossRef
172.
go back to reference Strasser A, Rolink A, Melchers F (1989) One synchronous wave of B cell development in mouse fetal liver changes at day 16 of gestation from dependence to independence of a stromal cell environment. J Exp Med 170(6):1973–1986PubMedCrossRef Strasser A, Rolink A, Melchers F (1989) One synchronous wave of B cell development in mouse fetal liver changes at day 16 of gestation from dependence to independence of a stromal cell environment. J Exp Med 170(6):1973–1986PubMedCrossRef
173.
go back to reference Paige CJ, Kincade PW, Shinefeld LA, Sato VL (1981) Precursors of murine B lymphocytes. Physical and functional characterization, and distinctions from myeloid stem cells. J Exp Med 153(1):154–165PubMedCrossRef Paige CJ, Kincade PW, Shinefeld LA, Sato VL (1981) Precursors of murine B lymphocytes. Physical and functional characterization, and distinctions from myeloid stem cells. J Exp Med 153(1):154–165PubMedCrossRef
174.
go back to reference Lekva T, Bollerslev J, Kristo C, Olstad OK, Ueland T, Jemtland R (2009) The glucocorticoid-induced leucine zipper gene (GILZ) expression decreases after successful treatment of patients with endogenous Cushing’s syndrome and may play a role in glucocorticoid-induced osteoporosis. J Clin Endocrinol Metab 95(1):246–255PubMedCrossRef Lekva T, Bollerslev J, Kristo C, Olstad OK, Ueland T, Jemtland R (2009) The glucocorticoid-induced leucine zipper gene (GILZ) expression decreases after successful treatment of patients with endogenous Cushing’s syndrome and may play a role in glucocorticoid-induced osteoporosis. J Clin Endocrinol Metab 95(1):246–255PubMedCrossRef
175.
go back to reference Lindton B, Markling L, Ringden O, Westgren M (2002) In vitro studies of haematopoietic colony-forming capacity of human fetal liver cells at exposure to cytotoxic and immunomodulatory drugs. Fetal Diagn Ther 17(2):104–109PubMedCrossRef Lindton B, Markling L, Ringden O, Westgren M (2002) In vitro studies of haematopoietic colony-forming capacity of human fetal liver cells at exposure to cytotoxic and immunomodulatory drugs. Fetal Diagn Ther 17(2):104–109PubMedCrossRef
176.
go back to reference Flygare J, Rayon Estrada V, Shin C, Gupta S, Lodish HF (2011) HIF1alpha synergizes with glucocorticoids to promote BFU-E progenitor self-renewal. Blood 117(12):3435–3444PubMedPubMedCentralCrossRef Flygare J, Rayon Estrada V, Shin C, Gupta S, Lodish HF (2011) HIF1alpha synergizes with glucocorticoids to promote BFU-E progenitor self-renewal. Blood 117(12):3435–3444PubMedPubMedCentralCrossRef
177.
go back to reference Lee HY, Gao X, Barrasa MI, Li H, Elmes RR, Peters LL, Lodish HF (2015) PPAR-alpha and glucocorticoid receptor synergize to promote erythroid progenitor self-renewal. Nature 522(7557):474–477PubMedPubMedCentralCrossRef Lee HY, Gao X, Barrasa MI, Li H, Elmes RR, Peters LL, Lodish HF (2015) PPAR-alpha and glucocorticoid receptor synergize to promote erythroid progenitor self-renewal. Nature 522(7557):474–477PubMedPubMedCentralCrossRef
178.
go back to reference Elahi S, Ertelt JM, Kinder JM, Jiang TT, Zhang X, Xin L, Chaturvedi V, Strong BS, Qualls JE, Steinbrecher KA, Kalfa TA, Shaaban AF, Way SS (2013) Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection. Nature 504(7478):158–162PubMedPubMedCentralCrossRef Elahi S, Ertelt JM, Kinder JM, Jiang TT, Zhang X, Xin L, Chaturvedi V, Strong BS, Qualls JE, Steinbrecher KA, Kalfa TA, Shaaban AF, Way SS (2013) Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection. Nature 504(7478):158–162PubMedPubMedCentralCrossRef
179.
go back to reference Sobrian SK, Vaughn VT, Ashe WK, Markovic B, Djuric V, Jankovic BD (1997) Gestational exposure to loud noise alters the development and postnatal responsiveness of humoral and cellular components of the immune system in offspring. Environ Res 73(1–2):227–241. doi:10.1006/enrs.1997.3734 PubMedCrossRef Sobrian SK, Vaughn VT, Ashe WK, Markovic B, Djuric V, Jankovic BD (1997) Gestational exposure to loud noise alters the development and postnatal responsiveness of humoral and cellular components of the immune system in offspring. Environ Res 73(1–2):227–241. doi:10.​1006/​enrs.​1997.​3734 PubMedCrossRef
180.
go back to reference Llorente E, Brito ML, Machado P, Gonzalez MC (2002) Effect of prenatal stress on the hormonal response to acute and chronic stress and on immune parameters in the offspring. J Physiol Biochem 58(3):143–149PubMedCrossRef Llorente E, Brito ML, Machado P, Gonzalez MC (2002) Effect of prenatal stress on the hormonal response to acute and chronic stress and on immune parameters in the offspring. J Physiol Biochem 58(3):143–149PubMedCrossRef
181.
go back to reference Mizoguchi T, Pinho S, Ahmed J, Kunisaki Y, Hanoun M, Mendelson A, Ono N, Kronenberg HM, Frenette PS (2014) Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev Cell 29(3):340–349PubMedPubMedCentralCrossRef Mizoguchi T, Pinho S, Ahmed J, Kunisaki Y, Hanoun M, Mendelson A, Ono N, Kronenberg HM, Frenette PS (2014) Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev Cell 29(3):340–349PubMedPubMedCentralCrossRef
182.
go back to reference Li A, Hardy R, Stoner S, Tuckermann J, Seibel M, Zhou H (2013) Deletion of mesenchymal glucocorticoid receptor attenuates embryonic lung development and abdominal wall closure. PLoS ONE 8(5), e63578PubMedPubMedCentralCrossRef Li A, Hardy R, Stoner S, Tuckermann J, Seibel M, Zhou H (2013) Deletion of mesenchymal glucocorticoid receptor attenuates embryonic lung development and abdominal wall closure. PLoS ONE 8(5), e63578PubMedPubMedCentralCrossRef
183.
go back to reference Korakaki E, Gourgiotis D, Aligizakis A, Manoura A, Hatzidaki E, Giahnakis E, Marmarinos A, Kalmanti M, Giannakopoulou C (2007) Levels of bone collagen markers in preterm infants: relation to antenatal glucocorticoid treatment. J Bone Miner Metab 25(3):172–178PubMedCrossRef Korakaki E, Gourgiotis D, Aligizakis A, Manoura A, Hatzidaki E, Giahnakis E, Marmarinos A, Kalmanti M, Giannakopoulou C (2007) Levels of bone collagen markers in preterm infants: relation to antenatal glucocorticoid treatment. J Bone Miner Metab 25(3):172–178PubMedCrossRef
184.
go back to reference Fonseca L, Ramin SM, Mele L, Wapner RJ, Johnson F, Peaceman AM, Sorokin Y, Dudley DJ, Spong CY, Leveno KJ, Caritis SN, Miodovnik M, Mercer B, Thorp JM, O’Sullivan MJ, Carpenter MW, Rouse DJ, Sibai B (2009) Bone metabolism in fetuses of pregnant women exposed to single and multiple courses of corticosteroids. Obstet Gynecol 114(1):38–44PubMedPubMedCentralCrossRef Fonseca L, Ramin SM, Mele L, Wapner RJ, Johnson F, Peaceman AM, Sorokin Y, Dudley DJ, Spong CY, Leveno KJ, Caritis SN, Miodovnik M, Mercer B, Thorp JM, O’Sullivan MJ, Carpenter MW, Rouse DJ, Sibai B (2009) Bone metabolism in fetuses of pregnant women exposed to single and multiple courses of corticosteroids. Obstet Gynecol 114(1):38–44PubMedPubMedCentralCrossRef
185.
go back to reference Vitale C, Cottalasso F, Montaldo E, Moretta L, Mingari MC (2008) Methylprednisolone induces preferential and rapid differentiation of CD34+ cord blood precursors toward NK cells. Int Immunol 20(4):565–575PubMedCrossRef Vitale C, Cottalasso F, Montaldo E, Moretta L, Mingari MC (2008) Methylprednisolone induces preferential and rapid differentiation of CD34+ cord blood precursors toward NK cells. Int Immunol 20(4):565–575PubMedCrossRef
187.
go back to reference Vacchio MS, Papadopoulos V, Ashwell JD (1994) Steroid production in the thymus: implications for thymocyte selection. J Exp Med 179(6):1835–1846PubMedCrossRef Vacchio MS, Papadopoulos V, Ashwell JD (1994) Steroid production in the thymus: implications for thymocyte selection. J Exp Med 179(6):1835–1846PubMedCrossRef
188.
go back to reference Taves MD, Plumb AW, Sandkam BA, Ma C, Van Der Gugten JG, Holmes DT, Close DA, Abraham N, Soma KK (2015) Steroid profiling reveals widespread local regulation of glucocorticoid levels during mouse development. Endocrinology 156(2):511–522. doi:10.1210/en.2013-1606 PubMedCrossRef Taves MD, Plumb AW, Sandkam BA, Ma C, Van Der Gugten JG, Holmes DT, Close DA, Abraham N, Soma KK (2015) Steroid profiling reveals widespread local regulation of glucocorticoid levels during mouse development. Endocrinology 156(2):511–522. doi:10.​1210/​en.​2013-1606 PubMedCrossRef
189.
go back to reference Wyllie AH (1980) Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature 284(5756):555–556PubMedCrossRef Wyllie AH (1980) Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature 284(5756):555–556PubMedCrossRef
192.
go back to reference Tolosa E, King LB, Ashwell JD (1998) Thymocyte glucocorticoid resistance alters positive selection and inhibits autoimmunity and lymphoproliferative disease in MRL-lpr/lpr mice. Immunity 8(1):67–76PubMedCrossRef Tolosa E, King LB, Ashwell JD (1998) Thymocyte glucocorticoid resistance alters positive selection and inhibits autoimmunity and lymphoproliferative disease in MRL-lpr/lpr mice. Immunity 8(1):67–76PubMedCrossRef
193.
go back to reference Lu FW, Yasutomo K, Goodman GB, McHeyzer-Williams LJ, McHeyzer-Williams MG, Germain RN, Ashwell JD (2000) Thymocyte resistance to glucocorticoids leads to antigen-specific unresponsiveness due to “holes” in the T cell repertoire. Immunity 12(2):183–192PubMedCrossRef Lu FW, Yasutomo K, Goodman GB, McHeyzer-Williams LJ, McHeyzer-Williams MG, Germain RN, Ashwell JD (2000) Thymocyte resistance to glucocorticoids leads to antigen-specific unresponsiveness due to “holes” in the T cell repertoire. Immunity 12(2):183–192PubMedCrossRef
195.
go back to reference Cole TJ, Myles K, Purton JF, Brereton PS, Solomon NM, Godfrey DI, Funder JW (2001) GRKO mice express an aberrant dexamethasone-binding glucocorticoid receptor, but are profoundly glucocorticoid resistant. Mol Cell Endocrinol 173(1–2):193–202PubMedCrossRef Cole TJ, Myles K, Purton JF, Brereton PS, Solomon NM, Godfrey DI, Funder JW (2001) GRKO mice express an aberrant dexamethasone-binding glucocorticoid receptor, but are profoundly glucocorticoid resistant. Mol Cell Endocrinol 173(1–2):193–202PubMedCrossRef
197.
go back to reference Purton JF, Boyd RL, Cole TJ, Godfrey DI (2000) Intrathymic T cell development and selection proceeds normally in the absence of glucocorticoid receptor signaling. Immunity 13(2):179–186PubMedCrossRef Purton JF, Boyd RL, Cole TJ, Godfrey DI (2000) Intrathymic T cell development and selection proceeds normally in the absence of glucocorticoid receptor signaling. Immunity 13(2):179–186PubMedCrossRef
199.
go back to reference Liddicoat DR, Purton JF, Cole TJ, Godfrey DI (2014) Glucocorticoid-mediated repression of T-cell receptor signalling is impaired in glucocorticoid receptor exon 2-disrupted mice. Immunol Cell Biol 92(2):148–155. doi:10.1038/icb.2013.76 PubMedCrossRef Liddicoat DR, Purton JF, Cole TJ, Godfrey DI (2014) Glucocorticoid-mediated repression of T-cell receptor signalling is impaired in glucocorticoid receptor exon 2-disrupted mice. Immunol Cell Biol 92(2):148–155. doi:10.​1038/​icb.​2013.​76 PubMedCrossRef
200.
go back to reference Diepenbruck I, Much CC, Krumbholz A, Kolster M, Thieme R, Thieme D, Diepenbruck S, Solano ME, Arck PC, Tolosa E (2013) Effect of prenatal steroid treatment on the developing immune system. J Mol Med (Berl) 91(11):1293–1302. doi:10.1007/s00109-013-1069-2 CrossRef Diepenbruck I, Much CC, Krumbholz A, Kolster M, Thieme R, Thieme D, Diepenbruck S, Solano ME, Arck PC, Tolosa E (2013) Effect of prenatal steroid treatment on the developing immune system. J Mol Med (Berl) 91(11):1293–1302. doi:10.​1007/​s00109-013-1069-2 CrossRef
201.
go back to reference Hartwig IR, Sly PD, Schmidt LA, van Lieshout RJ, Bienenstock J, Holt PG, Arck PC (2014) Prenatal adverse life events increase the risk for atopic diseases in children, which is enhanced in the absence of a maternal atopic predisposition. J Allergy Clin Immunol 134(1):160–169PubMedCrossRef Hartwig IR, Sly PD, Schmidt LA, van Lieshout RJ, Bienenstock J, Holt PG, Arck PC (2014) Prenatal adverse life events increase the risk for atopic diseases in children, which is enhanced in the absence of a maternal atopic predisposition. J Allergy Clin Immunol 134(1):160–169PubMedCrossRef
202.
go back to reference D’Adamio F, Zollo O, Moraca R, Ayroldi E, Bruscoli S, Bartoli A, Cannarile L, Migliorati G, Riccardi C (1997) A new dexamethasone-induced gene of the leucine zipper family protects T lymphocytes from TCR/CD3-activated cell death. Immunity 7(6):803–812PubMedCrossRef D’Adamio F, Zollo O, Moraca R, Ayroldi E, Bruscoli S, Bartoli A, Cannarile L, Migliorati G, Riccardi C (1997) A new dexamethasone-induced gene of the leucine zipper family protects T lymphocytes from TCR/CD3-activated cell death. Immunity 7(6):803–812PubMedCrossRef
204.
go back to reference Delfino DV, Agostini M, Spinicelli S, Vacca C, Riccardi C (2006) Inhibited cell death, NF-kappaB activity and increased IL-10 in TCR-triggered thymocytes of transgenic mice overexpressing the glucocorticoid-induced protein GILZ. Int Immunopharmacol 6(7):1126–1134. doi:10.1016/j.intimp.2006.02.001 PubMedCrossRef Delfino DV, Agostini M, Spinicelli S, Vacca C, Riccardi C (2006) Inhibited cell death, NF-kappaB activity and increased IL-10 in TCR-triggered thymocytes of transgenic mice overexpressing the glucocorticoid-induced protein GILZ. Int Immunopharmacol 6(7):1126–1134. doi:10.​1016/​j.​intimp.​2006.​02.​001 PubMedCrossRef
205.
go back to reference Calmette J, Ellouze M, Tran T, Karaki S, Ronin E, Capel F, Pallardy M, Bachelerie F, Krzysiek R, Emilie D, Schlecht-Louf G, Godot V (2014) Glucocorticoid-induced leucine zipper enhanced expression in dendritic cells is sufficient to drive regulatory T cells expansion in vivo. J Immunol 193(12):5863–5872. doi:10.4049/jimmunol.1400758 PubMedCrossRef Calmette J, Ellouze M, Tran T, Karaki S, Ronin E, Capel F, Pallardy M, Bachelerie F, Krzysiek R, Emilie D, Schlecht-Louf G, Godot V (2014) Glucocorticoid-induced leucine zipper enhanced expression in dendritic cells is sufficient to drive regulatory T cells expansion in vivo. J Immunol 193(12):5863–5872. doi:10.​4049/​jimmunol.​1400758 PubMedCrossRef
206.
go back to reference Bereshchenko O, Coppo M, Bruscoli S, Biagioli M, Cimino M, Frammartino T, Sorcini D, Venanzi A, Di Sante M, Riccardi C (2014) GILZ promotes production of peripherally induced Treg cells and mediates the crosstalk between glucocorticoids and TGF-beta signaling. Cell Rep 7(2):464–475. doi:10.1016/j.celrep.2014.03.004 PubMedCrossRef Bereshchenko O, Coppo M, Bruscoli S, Biagioli M, Cimino M, Frammartino T, Sorcini D, Venanzi A, Di Sante M, Riccardi C (2014) GILZ promotes production of peripherally induced Treg cells and mediates the crosstalk between glucocorticoids and TGF-beta signaling. Cell Rep 7(2):464–475. doi:10.​1016/​j.​celrep.​2014.​03.​004 PubMedCrossRef
207.
go back to reference Bruscoli S, Biagioli M, Sorcini D, Frammartino T, Cimino M, Sportoletti P, Mazzon E, Bereshchenko O, Riccardi C (2015) Lack of glucocorticoid-induced leucine zipper (GILZ) deregulates B-cell survival and results in B-cell lymphocytosis in mice. Blood 126(15):1790–1801. doi:10.1182/blood-2015-03-631580 PubMedPubMedCentralCrossRef Bruscoli S, Biagioli M, Sorcini D, Frammartino T, Cimino M, Sportoletti P, Mazzon E, Bereshchenko O, Riccardi C (2015) Lack of glucocorticoid-induced leucine zipper (GILZ) deregulates B-cell survival and results in B-cell lymphocytosis in mice. Blood 126(15):1790–1801. doi:10.​1182/​blood-2015-03-631580 PubMedPubMedCentralCrossRef
208.
209.
go back to reference Philips A, Maira M, Mullick A, Chamberland M, Lesage S, Hugo P, Drouin J (1997) Antagonism between Nur77 and glucocorticoid receptor for control of transcription. Mol Cell Biol 17(10):5952–5959PubMedPubMedCentralCrossRef Philips A, Maira M, Mullick A, Chamberland M, Lesage S, Hugo P, Drouin J (1997) Antagonism between Nur77 and glucocorticoid receptor for control of transcription. Mol Cell Biol 17(10):5952–5959PubMedPubMedCentralCrossRef
210.
211.
go back to reference Zhou T, Cheng J, Yang P, Wang Z, Liu C, Su X, Bluethmann H, Mountz JD (1996) Inhibition of Nur77/Nurr1 leads to inefficient clonal deletion of self-reactive T cells. J Exp Med 183(4):1879–1892PubMedCrossRef Zhou T, Cheng J, Yang P, Wang Z, Liu C, Su X, Bluethmann H, Mountz JD (1996) Inhibition of Nur77/Nurr1 leads to inefficient clonal deletion of self-reactive T cells. J Exp Med 183(4):1879–1892PubMedCrossRef
212.
go back to reference Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298(5597):1395–1401. doi:10.1126/science.1075958 PubMedCrossRef Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298(5597):1395–1401. doi:10.​1126/​science.​1075958 PubMedCrossRef
215.
go back to reference Fletcher AL, Lowen TE, Sakkal S, Reiseger JJ, Hammett MV, Seach N, Scott HS, Boyd RL, Chidgey AP (2009) Ablation and regeneration of tolerance-inducing medullary thymic epithelial cells after cyclosporine, cyclophosphamide, and dexamethasone treatment. J Immunol 183(2):823–831. doi:10.4049/jimmunol.0900225 PubMedCrossRef Fletcher AL, Lowen TE, Sakkal S, Reiseger JJ, Hammett MV, Seach N, Scott HS, Boyd RL, Chidgey AP (2009) Ablation and regeneration of tolerance-inducing medullary thymic epithelial cells after cyclosporine, cyclophosphamide, and dexamethasone treatment. J Immunol 183(2):823–831. doi:10.​4049/​jimmunol.​0900225 PubMedCrossRef
216.
go back to reference Drake AJ, Seckl JR (2012) Transmission of programming effects across generations. Pediatr Endocrinol Rev 9:566–578 Drake AJ, Seckl JR (2012) Transmission of programming effects across generations. Pediatr Endocrinol Rev 9:566–578
217.
go back to reference Meaney MJ, Ferguson-Smith AC (2010) Epigenetic regulation of the neural transcriptome: the meaning of the marks. Nat Neurosci 13(11):1313–1318PubMedCrossRef Meaney MJ, Ferguson-Smith AC (2010) Epigenetic regulation of the neural transcriptome: the meaning of the marks. Nat Neurosci 13(11):1313–1318PubMedCrossRef
218.
go back to reference Moisiadis VG, Matthews SG (2014) Glucocorticoids and fetal programming part 2: mechanisms. Nat Rev Endocrinol 10(7):403–411PubMedCrossRef Moisiadis VG, Matthews SG (2014) Glucocorticoids and fetal programming part 2: mechanisms. Nat Rev Endocrinol 10(7):403–411PubMedCrossRef
219.
220.
go back to reference Oh IH, Humphries RK (2012) Concise review: multidimensional regulation of the hematopoietic stem cell state. Stem Cells 30(1):82–88PubMedCrossRef Oh IH, Humphries RK (2012) Concise review: multidimensional regulation of the hematopoietic stem cell state. Stem Cells 30(1):82–88PubMedCrossRef
221.
go back to reference Taiwo O, Wilson GA, Emmett W, Morris T, Bonnet D, Schuster E, Adejumo T, Beck S, Pearce DJ (2013) DNA methylation analysis of murine hematopoietic side population cells during aging. Epigenetics 8(10):1114–1122PubMedPubMedCentralCrossRef Taiwo O, Wilson GA, Emmett W, Morris T, Bonnet D, Schuster E, Adejumo T, Beck S, Pearce DJ (2013) DNA methylation analysis of murine hematopoietic side population cells during aging. Epigenetics 8(10):1114–1122PubMedPubMedCentralCrossRef
222.
go back to reference Ji H, Ehrlich LI, Seita J, Murakami P, Doi A, Lindau P, Lee H, Aryee MJ, Irizarry RA, Kim K, Rossi DJ, Inlay MA, Serwold T, Karsunky H, Ho L, Daley GQ, Weissman IL, Feinberg AP (2010) Comprehensive methylome map of lineage commitment from haematopoietic progenitors. Nature 467(7313):338–342PubMedPubMedCentralCrossRef Ji H, Ehrlich LI, Seita J, Murakami P, Doi A, Lindau P, Lee H, Aryee MJ, Irizarry RA, Kim K, Rossi DJ, Inlay MA, Serwold T, Karsunky H, Ho L, Daley GQ, Weissman IL, Feinberg AP (2010) Comprehensive methylome map of lineage commitment from haematopoietic progenitors. Nature 467(7313):338–342PubMedPubMedCentralCrossRef
223.
go back to reference Broske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R, Nerlov C, Leutz A, Andrade-Navarro MA, Jacobsen SE, Rosenbauer F (2009) DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet 41(11):1207–1215. doi:10.1038/ng.463 PubMedCrossRef Broske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R, Nerlov C, Leutz A, Andrade-Navarro MA, Jacobsen SE, Rosenbauer F (2009) DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet 41(11):1207–1215. doi:10.​1038/​ng.​463 PubMedCrossRef
225.
go back to reference Bartholdy B, Christopeit M, Will B, Mo Y, Barreyro L, Yu Y, Bhagat TD, Okoye-Okafor UC, Todorova TI, Greally JM, Levine RL, Melnick A, Verma A, Steidl U (2014) HSC commitment-associated epigenetic signature is prognostic in acute myeloid leukemia. J Clin Invest 124(3):1158–1167PubMedPubMedCentralCrossRef Bartholdy B, Christopeit M, Will B, Mo Y, Barreyro L, Yu Y, Bhagat TD, Okoye-Okafor UC, Todorova TI, Greally JM, Levine RL, Melnick A, Verma A, Steidl U (2014) HSC commitment-associated epigenetic signature is prognostic in acute myeloid leukemia. J Clin Invest 124(3):1158–1167PubMedPubMedCentralCrossRef
226.
go back to reference McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonte B, Szyf M, Turecki G, Meaney MJ (2009) Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci 12(3):342–348PubMedPubMedCentralCrossRef McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonte B, Szyf M, Turecki G, Meaney MJ (2009) Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci 12(3):342–348PubMedPubMedCentralCrossRef
227.
go back to reference Petropoulos S, Matthews SG, Szyf M (2014) Adult glucocorticoid exposure leads to transcriptional and DNA methylation changes in nuclear steroid receptors in the hippocampus and kidney of mouse male offspring. Biol Reprod 90(2):43PubMedCrossRef Petropoulos S, Matthews SG, Szyf M (2014) Adult glucocorticoid exposure leads to transcriptional and DNA methylation changes in nuclear steroid receptors in the hippocampus and kidney of mouse male offspring. Biol Reprod 90(2):43PubMedCrossRef
228.
go back to reference Rodriguez RM, Suarez-Alvarez B, Mosen-Ansorena D, Garcia-Peydro M, Fuentes P, Garcia-Leon MJ, Gonzalez-Lahera A, Macias-Camara N, Toribio ML, Aransay AM, Lopez-Larrea C (2015) Regulation of the transcriptional program by DNA methylation during human alphabeta T-cell development. Nucleic Acids Res 43(2):760–774. doi:10.1093/nar/gku1340 PubMedCrossRef Rodriguez RM, Suarez-Alvarez B, Mosen-Ansorena D, Garcia-Peydro M, Fuentes P, Garcia-Leon MJ, Gonzalez-Lahera A, Macias-Camara N, Toribio ML, Aransay AM, Lopez-Larrea C (2015) Regulation of the transcriptional program by DNA methylation during human alphabeta T-cell development. Nucleic Acids Res 43(2):760–774. doi:10.​1093/​nar/​gku1340 PubMedCrossRef
230.
go back to reference Sellars M, Huh JR, Day K, Issuree PD, Galan C, Gobeil S, Absher D, Green MR, Littman DR (2015) Regulation of DNA methylation dictates Cd4 expression during the development of helper and cytotoxic T cell lineages. Nat Immunol 16(7):746–754. doi:10.1038/ni.3198 PubMedPubMedCentralCrossRef Sellars M, Huh JR, Day K, Issuree PD, Galan C, Gobeil S, Absher D, Green MR, Littman DR (2015) Regulation of DNA methylation dictates Cd4 expression during the development of helper and cytotoxic T cell lineages. Nat Immunol 16(7):746–754. doi:10.​1038/​ni.​3198 PubMedPubMedCentralCrossRef
234.
go back to reference Galon J, Franchimont D, Hiroi N, Frey G, Boettner A, Ehrhart-Bornstein M, O’Shea JJ, Chrousos GP, Bornstein SR (2002) Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J 16(1):61–71. doi:10.1096/fj.01-0245com16/1/61 PubMedCrossRef Galon J, Franchimont D, Hiroi N, Frey G, Boettner A, Ehrhart-Bornstein M, O’Shea JJ, Chrousos GP, Bornstein SR (2002) Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J 16(1):61–71. doi:10.​1096/​fj.​01-0245com16/​1/​61 PubMedCrossRef
237.
go back to reference Cannarile L, Fallarino F, Agostini M, Cuzzocrea S, Mazzon E, Vacca C, Genovese T, Migliorati G, Ayroldi E, Riccardi C (2006) Increased GILZ expression in transgenic mice up-regulates Th-2 lymphokines. Blood 107(3):1039–1047. doi:10.1182/blood-2005-05-2183 PubMedCrossRef Cannarile L, Fallarino F, Agostini M, Cuzzocrea S, Mazzon E, Vacca C, Genovese T, Migliorati G, Ayroldi E, Riccardi C (2006) Increased GILZ expression in transgenic mice up-regulates Th-2 lymphokines. Blood 107(3):1039–1047. doi:10.​1182/​blood-2005-05-2183 PubMedCrossRef
238.
go back to reference Lu Y, Ho R, Lim TK, Kuan WS, Goh DY, Mahadevan M, Sim TB, Van Bever HP, Larbi A, Ng TP (2015) Neuropeptide Y may mediate psychological stress and enhance TH2 inflammatory response in asthma. J Allergy Clin Immunol 135(4):1061–1063 e1064. doi:10.1016/j.jaci.2014.10.036 PubMedCrossRef Lu Y, Ho R, Lim TK, Kuan WS, Goh DY, Mahadevan M, Sim TB, Van Bever HP, Larbi A, Ng TP (2015) Neuropeptide Y may mediate psychological stress and enhance TH2 inflammatory response in asthma. J Allergy Clin Immunol 135(4):1061–1063 e1064. doi:10.​1016/​j.​jaci.​2014.​10.​036 PubMedCrossRef
240.
go back to reference Knight DA, Holgate ST (2003) The airway epithelium: structural and functional properties in health and disease. Respirology 8(4):432–446PubMedCrossRef Knight DA, Holgate ST (2003) The airway epithelium: structural and functional properties in health and disease. Respirology 8(4):432–446PubMedCrossRef
241.
go back to reference Holgate ST, Lackie PM, Howarth PH, Roche WR, Puddicombe SM, Richter A, Wilson SJ, Holloway JW, Davies DE (2001) Invited lecture: activation of the epithelial mesenchymal trophic unit in the pathogenesis of asthma. Int Arch Allergy Immunol 124(1–3):253–258PubMed Holgate ST, Lackie PM, Howarth PH, Roche WR, Puddicombe SM, Richter A, Wilson SJ, Holloway JW, Davies DE (2001) Invited lecture: activation of the epithelial mesenchymal trophic unit in the pathogenesis of asthma. Int Arch Allergy Immunol 124(1–3):253–258PubMed
242.
go back to reference Bird AD, Choo YL, Hooper SB, McDougall AR, Cole TJ (2014) Mesenchymal glucocorticoid receptor regulates the development of multiple cell layers of the mouse lung. Am J Respir Cell Mol Biol 50(2):419–428. doi:10.1165/rcmb.2013-0169OC PubMed Bird AD, Choo YL, Hooper SB, McDougall AR, Cole TJ (2014) Mesenchymal glucocorticoid receptor regulates the development of multiple cell layers of the mouse lung. Am J Respir Cell Mol Biol 50(2):419–428. doi:10.​1165/​rcmb.​2013-0169OC PubMed
244.
go back to reference Crompton R, Clifton VL, Bisits AT, Read MA, Smith R, Wright IM (2003) Corticotropin-releasing hormone causes vasodilation in human skin via mast cell-dependent pathways. J Clin Endocrinol Metab 88(11):5427–5432. doi:10.1210/jc.2003-030377 PubMedCrossRef Crompton R, Clifton VL, Bisits AT, Read MA, Smith R, Wright IM (2003) Corticotropin-releasing hormone causes vasodilation in human skin via mast cell-dependent pathways. J Clin Endocrinol Metab 88(11):5427–5432. doi:10.​1210/​jc.​2003-030377 PubMedCrossRef
245.
go back to reference Theoharides TC, Cochrane DE (2004) Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 146(1–2):1–12PubMedCrossRef Theoharides TC, Cochrane DE (2004) Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 146(1–2):1–12PubMedCrossRef
246.
248.
go back to reference Steffensen FH, Sorensen HT, Gillman MW, Rothman KJ, Sabroe S, Fischer P, Olsen J (2000) Low birth weight and preterm delivery as risk factors for asthma and atopic dermatitis in young adult males. Epidemiology 11(2):185–188PubMedCrossRef Steffensen FH, Sorensen HT, Gillman MW, Rothman KJ, Sabroe S, Fischer P, Olsen J (2000) Low birth weight and preterm delivery as risk factors for asthma and atopic dermatitis in young adult males. Epidemiology 11(2):185–188PubMedCrossRef
Metadata
Title
Antenatal endogenous and exogenous glucocorticoids and their impact on immune ontogeny and long-term immunity
Authors
María Emilia Solano
Megan C. Holmes
Paul R. Mittelstadt
Karen E. Chapman
Eva Tolosa
Publication date
01-11-2016
Publisher
Springer Berlin Heidelberg
Published in
Seminars in Immunopathology / Issue 6/2016
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-016-0575-z

Other articles of this Issue 6/2016

Seminars in Immunopathology 6/2016 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine