Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-26T07:10:28.466Z Has data issue: false hasContentIssue false

Section I - Hormones in the Physiology and Pharmacology of Pregnancy

Published online by Cambridge University Press:  09 November 2022

Felice Petraglia
Affiliation:
Università degli Studi, Florence
Mariarosaria Di Tommaso
Affiliation:
Università degli Studi, Florence
Federico Mecacci
Affiliation:
Università degli Studi, Florence
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Hormones and Pregnancy
Basic Science and Clinical Implications
, pp. 1 - 72
Publisher: Cambridge University Press
Print publication year: 2022

Access options

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

References

References

Vannuccini, S, Bocchi, C, Severi, FM, et al. Endocrinology of human parturition. Ann Endocrinol (Paris). 2016, 77(2):105113.Google Scholar
Voltolini, C, and Petraglia, F. Neuroendocrinology of pregnancy and parturition. In: Handbook of Clinical Neurology. 2014.CrossRefGoogle Scholar
Behura, SK, Dhakal, P, Kelleher, AM, et al. The brain-placental axis: Therapeutic and pharmacological relevancy to pregnancy. Pharmacological Research. 2019.Google Scholar
Petraglia, F, Imperatore, A, and Challis, JRG. Neuroendocrine mechanisms in pregnancy and parturition. Endocrine Reviews. 2010, 31: 783816.CrossRefGoogle ScholarPubMed
Iliodromiti, Z, Antonakopoulos, N, Sifakis, S, et al. Endocrine, paracrine, and autocrine placental mediators in labor. Hormones. 2012.Google Scholar
Petraglia, F, Florio, P, Nappi, C, et al. Peptide signaling in human placenta and membranes: Autocrine, paracrine, and endocrine mechanisms. Vol., Endocrine Reviews. 1996, 17: 156186.Google Scholar
Petraglia, F, Florio, P, and Torricelli, M. Placental endocrine function. In: Knobil and Neill’s Physiology of Reproduction. 2006.Google Scholar
Brunton, PJ, and Russell, JA. Endocrine induced changes in brain function during pregnancy. Brain Research. 2010.Google Scholar
Warren, WB, and Silverman, AJ. Cellular localization of corticotrophin releasing hormone in the human placenta, fetal membranes and decidua. Placenta. 1995.Google Scholar
Vaughan, J, Donaldson, C, Bittencourt, J, et al. Urocortin, a mammalian neuropeptide related to fish urotensin I and to corticotropin-releasing factor. Nature. 1995.Google Scholar
Zoumakis, E, Kalantaridou, S, and Makrigiannakis, A. CRH-like peptides in human reproduction. Curr Med Chem. 2009.Google Scholar
Welberg, LAM, and Seckl, JR. Prenatal stress, glucocorticoids and the programming of the brain. Journal of Neuroendocrinology. 2001.Google Scholar
Waddell, BJ. The placenta as hypothalamus and pituitary: Possible impact on maternal and fetal adrenal function. Reproduction, Fertility and Development. 1993.Google Scholar
Florio, P, Severi, FM, Ciarmela, P, et al. Placental stress factors and maternal-fetal adaptive response: The corticotropin-releasing factor family. Endocrine. 2002.Google Scholar
Challis, JRG, Bloomfield, FH, Booking, AD, et al. Fetal signals and parturition. Journal of Obstetrics and Gynaecology Research. 2005.Google Scholar
Ochȩdalski, T, Zylinńska, K, Laudański, T, et al. Corticotrophin-releasing hormone and ACTH levels in maternal and fetal blood during spontaneous and oxytocin-induced labour. European Journal of Endocrinology. 2001;Google Scholar
Vitoratos, N, Papatheodorou, DC, Kalantaridou, SN, et al. “Reproductive” corticotropin-releasing hormone. In: Annals of the New York Academy of Sciences. 2006.Google Scholar
Challis, JRG, Matthews, SG, Gibb, W, et al. Endocrine and paracrine regulation of birth at term and preterm. Endocrine Reviews. 2000.Google Scholar
McLean, M, Bisits, A, Davies, J, et al. A placental clock controlling the length of human pregnancy. Nature Medicine. 1995, 1(5):460463.CrossRefGoogle ScholarPubMed
Torricelli, M, Ignacchiti, E, Giovannelli, A, et al. Maternal plasma corticotrophin-releasing factor and urocortin levels in post-term pregnancies. European Journal of Endocrinology. 2006.Google Scholar
Rainey, WE, Rehman, KS, and Carr, BR. The human fetal adrenal: Making adrenal androgens for placental estrogens. Seminars in Reproductive Medicine. 2004.CrossRefGoogle Scholar
Smith, R, Smith, JI, Shen, X, et al. Patterns of plasma corticotropin-releasing hormone, progesterone, estradiol, and estriol change and the onset of human labor. Journal of Clinical Endocrinology & Metabolism. 2009, 94(6):20662074.Google Scholar
Grammatopoulos, DK. Placental corticotrophin-releasing hormone and its receptors in human pregnancy and labour: Still a scientific enigma. In: Journal of Neuroendocrinology. 2008.Google Scholar
McKeown, KJ, and Challis, JRG. Regulation of 15-hydroxy prostaglandin dehydrogenase by corticotrophin-releasing hormone through a calcium-dependent pathway in human chorion trophoblast cells. Journal of Clinical Endocrinology & Metabolism. 2003.Google Scholar
Grammatopoulos, DK, and Hillhouse, EW. Role of corticotropin-releasing hormone in onset of labour. Vol. 354, Lancet. 1999, 15461549.Google Scholar
Simpkin, JC, Kermani, F, Palmer, AM, et al. Effects of corticotrophin releasing hormone on contractile activity of myometrium from pregnant women. BJOG: An International Journal of Obstetrics & Gynaecology. 1999.Google Scholar
Jeschke, U, Mylonas, I, Richter, DU, et al. Regulation of progesterone production in human term trophoblasts in vitro by CRH, ACTH and cortisol (prednisolone). Archives of Gynecology and Obstetrics. 2005.CrossRefGoogle Scholar
Zhao, XJ, Hoheisel, C, Schauer, J, et al. Corticotropin-releasing hormone-binding protein and its possible role in neuroendocrinological research. Hormone and Metabolic Research. 1997.Google Scholar
Florio, P, Woods, RJ, Genazzani, AR, et al. Changes in amniotic fluid immunoreactive Corticotropin-Releasing Factor (CRF) and CRF-binding protein levels in pregnant women at term and during labor 1. Journal of Clinical Endocrinology & Metabolism. 1997.Google Scholar
Gitau, R, Fisk, NM, Teixeira, JMA, et al. Fetal hypothalamic-pituitary-adrenal stress responses to invasive procedures are independent of maternal responses. Journal of Clinical Endocrinology & Metabolism. 2001.Google Scholar
Mesiano, S, DeFranco, E, Muglia, LJ. Parturition. In: Knobil and Neill’s Physiology of Reproduction: Two-Volume Set. 2015.Google Scholar
Nepomnaschy, PA, Welch, KB, McConnell, DS, et al. Cortisol levels and very early pregnancy loss in humans. Proceedings of the National Academy of Sciences of the United States of America. 2006.Google Scholar
Hobel, CJ, Dunkel-Schetter, C, Roesch, SC, et al. Maternal plasma corticotropin-releasing hormone associated with stress at 20 weeks’ gestation in pregnancies ending in preterm delivery. American Journal of Obstetrics and Gynecology. 1999.Google Scholar
Torricelli, M, Novembri, R, Bloise, E, et al Changes in placental CRH, urocortins, and CRH-receptor mRNA expression associated with preterm delivery and chorioamnionitis. Journal of Clinical Endocrinology & Metabolism. 2011.Google Scholar
Florio, P, Imperatore, A, Sanseverino, F, et al. The measurement of maternal plasma corticotropin-releasing factor (CRF) and CRF-binding protein improves the early prediction of preeclampsia. Journal of Clinical Endocrinology & Metabolism. 2004.Google Scholar
Glynn, BP, Welton, A, Rodriguez-Linares, B, et al. Urocortin in pregnancy. American Journal of Obstetrics and Gynecology. 1998.Google Scholar
Petraglia, F, Florio, P, Benedetto, C, et al. Urocortin stimulates placental adrenocorticotropin and prostaglandin release and myometrial contractility in vitro. Journal of Clinical Endocrinology & Metabolism. 1999, 84(4):14201423.Google Scholar
Clifton, VL, Qing, G, Murphy, VE, et al. Localization and characterization of urocortin during human pregnancy. Placenta. 2000, 21(8):782788.CrossRefGoogle ScholarPubMed
Imperatore, A, Florio, P, Torres, PB, et al. Urocortin 2 and urocortin 3 are expressed by the human placenta, deciduas, and fetal membranes. Am Journal of Obstetrics and Gynaecology. 2006.CrossRefGoogle Scholar
Hashimoto, K, Nishiyama, M, Tanaka, Y, et al. Urocortins and corticotropin releasing factor type 2 receptors in the hypothalamus and the cardiovascular system. Peptides. 2004.Google Scholar
Karteris, E, Hillhouse, EW, and Grammatopoulos, D. Urocortin II Is expressed in human pregnant myometrial cells and regulates myosin light chain phosphorylation: Potential role of the type-2 Corticotropin-Releasing Hormone receptor in the control of myometrial contractility. Endocrinology. 2004;145(2):890900.Google Scholar
Florio, P, Torricelli, M, Galleri, L, et al. High fetal urocortin levels at term and preterm labor. Journal of Clinical Endocrinology & Metabolism. 2005.Google Scholar
Florio, P, Torricelli, M, De Falco, G, et al. High maternal and fetal plasma urocortin levels in pregnancies complicated by hypertension. Journal of Hypertension. 2006.Google Scholar
Imperatore, A, Rolfo, A, Petraglia, F, et al. Hypoxia and preeclampsia: Increased expression of urocortin 2 and urocortin 3. Reproductive Sciences. 2010.Google Scholar
Giovannelli, A, Greenwood, SL, Desforges, M, et al. Corticotrophin-releasing factor and urocortin inhibit system a activity in term human placental villous explants. Placenta. 2011.Google Scholar
Sasaki, K, and Norwitz, ER. Gonadotropin-releasing hormone/gonadotropin-releasing hormone receptor signaling in the placenta. Current Opinion in Endocrinology, Diabetes and Obesity. 2011.CrossRefGoogle Scholar
Lee, HJ, Snegovskikh, VV, Park, JS, et al. Role of GnRH-GnRH receptor signaling at the maternal-fetal interface. Fertility and Sterility. 2010.Google Scholar
Sassolas, G. Growth hormone-releasing hormone: Past and present. Hormone Research. 2000.Google Scholar
Frankenne, F, Scippo, ML, Van, BJ, et al. Identification of placental human growth hormone as the growth hormone-v gene expression product. Journal of Clinical Endocrinology & Metabolism. 1990.Google Scholar
Alsat, E, Guibourdenche, J, Couturier, A, et al. Physiological role of human placental growth hormone. Molecular and Cellular Endocrinology. 1998.Google Scholar
Davies, S, Byrn, F, and Cole, LA. Human chorionic gonadotropin testing for early pregnancy viability and complications. Clinics in Laboratory Medicine. 2003.Google Scholar
Williams, GR. Neurodevelopmental and neurophysiological actions of thyroid hormone. Journal of Neuroendocrinology. 2008.Google Scholar
Arrowsmith, S, and Wray, S. Oxytocin: Its mechanism of action and receptor signalling in the myometrium. Journal of Neuroendocrinology. 2014.Google Scholar
Uvnäs-Moberg, K, Ekström-Bergström, A, Berg, M, et al. Maternal plasma levels of oxytocin during physiological childbirth - A systematic review with implications for uterine contractions and central actions of oxytocin. BMC Pregnancy Childbirth. 2019.Google Scholar
Bossmar, T, Osman, N, Zilahi, E, et al. Expression of the oxytocin gene, but not the vasopressin gene, in the rat uterus during pregnancy: Influence of oestradiol and progesterone. Journal of Endocrinology. 2007.CrossRefGoogle Scholar
Fuchs, A‐R, and Fuchs, F. Endocrinology of human parturition: A review. BJOG An International Journal of Gynecology & Obstetrics. 1984.Google Scholar
Keverne, EB. Central mechanisms underlying the neural and neuroendocrine determinants of maternal behaviour. Psychoneuroendocrinology. 1988.Google Scholar
Freeman, ME, Kanyicska, B, Lerant, A, et al. Prolactin: Structure, function, and regulation of secretion. Physiological Reviews. 2000.Google Scholar
Ben-Jonathan, N, LaPensee, CR, and LaPensee, EW. What can we learn from rodents about prolactin in humans? Endocrine Reviews. 2008.Google Scholar
Grattan, DR, and Kokay, IC. Prolactin: A pleiotropic neuroendocrine hormone. Journal of Neuroendocrinology. 2008.Google Scholar
Walker, WH, Fitzpatrick, SL, Saunders, GF, et al. The human placental lactogen genes: Structure, function, evolution and transcriptional regulation. Endocrine Reviews. 1991.Google Scholar
Newbern, D, and Freemark, M. Placental hormones and the control of maternal metabolism and fetal growth. Current Opinion in Endocrinology, Diabetes and Obesity. 2011.Google Scholar
Sørensen, S, Von Tabouillot, D, Schiøler, V, et al. Serial measurements of serum human placental lactogen (hPL) and serial ultrasound examinations in the evaluation of fetal growth. Early Human Development. 2000.Google Scholar
Shingo, T, Gregg, C, Enwere, E, et al. Pregnancy-stimulated neurogenesis in the adult female forebrain mediated by prolactin. Science (80- ). 2003.Google Scholar
Torner, L, and Neumann, ID. The brain prolactin system: Involvement in stress response adaptations in lactation. Stress. 2002.Google Scholar
Sahay, A, Kale, A, and Joshi, S. Role of neurotrophins in pregnancy and offspring brain development. Neuropeptides. 2020.Google Scholar
Petraglia, F, Coukos, G, Battaglia, C, et al. Plasma and amniotic fluid immunoreactive neuropeptide-Y level changes during pregnancy, labor, and at parturition. Journal of Clinical Endocrinology & Metabolism. 1989.Google Scholar
Khatun, S, Kanayama, N, Belayet, HM, et al. Increased concentrations of plasma neuropeptide Y in patients with eclampsia and preeclampsia. American Journal of Obstetrics & Gynecology. 2000.Google Scholar
Marshall, SA, Senadheera, SN, Parry, LJ, et al. The role of relaxin in normal and abnormal uterine function during the menstrual cycle and early pregnancy. Reproductive Sciences. 2017.Google Scholar
Goldsmith, LT, Weiss, G, and Steinetz, BG. Relaxin and its role in pregnancy. Endocrinology and Metabolism Clinics of North America. 1995.Google Scholar
Clemens, TL, Cormier, S, Eichinger, A, et al. Parathyroid hormone-related protein and its receptors: Nuclear functions and roles in the renal and cardiovascular systems, the placental trophoblasts and the pancreatic islets. British Journal of Pharmacology. 2001.Google Scholar
Ferguson, JE, Gorman, JV, Bruns, DE, et al. Abundant expression of parathyroid hormone-related protein in human amnion and its association with labor. Proceedings of the National Academy of Sciences of the United States of America. 1992.Google Scholar
Ahmed, MS, Cemerikic, B, and Agbas, A. Properties and functions of human placental opioid system. Life Sciences. 1992.Google Scholar
Le Goascogne, C, Eychenne, B, Tonon, MC, et al. Neurosteroid progesterone is up-regulated in the brain of jimpy and shiverer mice. Glia. 2000.Google Scholar
Dombroski, RA, Casey, ML, and MacDonald, PC. 5α-dihydroprogesterone formation in human placenta from 5α-pregnan-3β/α-ol-20-ones and 5-pregnan-3β-yl-20-one sulfate. Journal of Steroid Biochemistry and Molecular Biology. 1997.Google Scholar
Luisi, S, Petraglia, F, Benedetto, C, et al. Serum allopregnanolone levels in pregnant women: Changes during pregnancy, at delivery, and in hypertensive patients. Journal of Clinical Endocrinology & Metabolism. 2000.Google Scholar
Manyonda, IT, Slater, DM, Fenske, C, et al. A role for noradrenaline in pre-eclampsia: Towards a unifying hypothesis for the pathophysiology. BJOG: An International Journal of Obstetrics & Gynaecology. 1998.Google Scholar
Nappi, RE, Petraglia, F, Luisi, S, et al. Serum allopregnanolone in women with postpartum “blues.” Obstetrics & Gynecology. 2001.Google Scholar
Pluchino, N, Ansaldi, Y, and Genazzani, AR. Brain intracrinology of allopregnanolone during pregnancy and hormonal contraception. Hormone Molecular Biology and Clinical Investigation. 2019.CrossRefGoogle Scholar

References

Smith, R, Paul, JW, and Tolosa, JM. Sharpey-Schafer Lecture 2019: From retroviruses to human birth. Exp Physiol. 2020, 105(4):555561.Google Scholar
Sultana, Z, Maiti, K, Aitken, J, et al. Oxidative stress, placental ageing-related pathologies and adverse pregnancy outcomes. Am J Reprod Immunol. 2017, 77(5).Google Scholar
Jin, J, and Menon, R. Placental exosomes: A proxy to understand pregnancy complications. Am J Reprod Immunol. 2018, 79(5):e12788.Google Scholar
Zeng, Y, and Chen, T. DNA methylation reprogramming during mammalian development. Genes (Basel). 2019, 10(4).Google Scholar
Tolosa, JM, Schjenken, JE, Clifton, VL, et al. The endogenous retroviral envelope protein syncytin-1 inhibits LPS/PHA-stimulated cytokine responses in human blood and is sorted into placental exosomes. Placenta. 2012, 33(11):933941.Google Scholar
Cheng, YH, and Handwerger, S. A placenta-specific enhancer of the human syncytin gene. Biol Reprod. 2005, 73(3):500509.Google Scholar
Makrigiannakis, A, Vrekoussis, T, Zoumakis, E, et al. The role of HCG in implantation: A mini-review of molecular and clinical evidence. Int J Mol Sci. 2017, 18(6).Google Scholar
Fisher, JJ, Bartho, LA, Perkins, AV, et al. Placental mitochondria and reactive oxygen species in the physiology and pathophysiology of pregnancy. Clin Exp Pharmacol Physiol. 2020, 47(1):176184.Google Scholar
Gomez-Concha, C, Flores-Herrera, O, Olvera-Sanchez, S, et al. Progesterone synthesis by human placental mitochondria is sensitive to PKA inhibition by H89. Int. J. Biochem. Cell Biol. 2011, 43(9):14021411.Google Scholar
Martinez, F, Olvera-Sanchez, S, Esparza-Perusquia, M, et al. Multiple functions of syncytiotrophoblast mitochondria. Steroids. 2015, 103:1122.Google Scholar
Berkane, N, Liere, P, Oudinet, JP, et al. From pregnancy to preeclampsia: A key role for estrogens. Endocr Rev. 2017, 38(2):123144.Google Scholar
Smith, R, Mesiano, S, Chan, EC, et al. Corticotropin-releasing hormone directly and preferentially stimulates dehydroepiandrosterone sulfate secretion by human fetal adrenal cortical cells. J Clin Endocrinol Metab. 1998, 83(8):29162920.Google Scholar
Nagashima, K, Yagi, H, Yunoki, H, et al. Cord blood levels of corticotropin-releasing factor. Biol Neonate. 1987, 51(1):14.Google Scholar
Clark, D, Thody, AJ, Shuster, S, et al. Immunoreactive alpha-MSH in human plasma in pregnancy. Nature. 1978, 273(5658):163164.CrossRefGoogle ScholarPubMed
Sasaki, A, Shinkawa, O, and Yoshinaga, K. Placental corticotropin-releasing hormone may be a stimulator of maternal pituitary adrenocorticotropic hormone secretion in humans. J Clin Invest. 1989;84(6), 19972001.Google Scholar
Steine, IM, LeWinn, KZ, Lisha, N, et al. Maternal exposure to childhood traumatic events, but not multi-domain psychosocial stressors, predict placental corticotrophin releasing hormone across pregnancy. Soc Sci Med. 2020, 266:113461.Google Scholar
Smith, R, Smith, JI, Shen, X, et al. Patterns of plasma corticotropin-releasing hormone, progesterone, estradiol, and estriol change and the onset of human labor. J Clin Endocrinol Metab. 2009, 94(6):20662074.Google Scholar
Melamed, M, Castano, E, Notides, AC, et al. Molecular and kinetic basis for the mixed agonist/antagonist activity of estriol. Mol Endocrinol. 1997, 11(12):18681878.Google Scholar
Lappano, R, Rosano, C, De Marco, P, et al. Estriol acts as a GPR30 antagonist in estrogen receptor-negative breast cancer cells. Mol Cell Endocrinol. 2010, 320(1-2):162170.Google Scholar
King, BR, Smith, R, and Nicholson, RC. Novel glucocorticoid and cAMP interactions on the CRH gene promoter. Mol Cell Endocrinol. 2002, 194(1–2), 1928.Google Scholar
Matsuo, H, Maruo, T, Hoshina, M, et al. [Selective inhibition of synthesis and secretion of hCG by progesterone and its correlation with hCG (alpha, beta) mRNA levels]. Nihon Naibunpi Gakkai Zasshi. 1985, 61(9):882892.Google Scholar
Morrish, DW, Marusyk, H, and Siy, O. Demonstration of specific secretory granules for human chorionic gonadotropin in placenta. J Histochem Cytochem. 1987, 35(1):93101.Google Scholar
Gellersen, B, Brosens, IA, and Brosens, JJ. Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Semin Reprod Med. 2007, 25(6):445453.Google Scholar
Burton, GJ, Jauniaux, E, and Charnock-Jones, DS. Human early placental development: potential roles of the endometrial glands. Placenta. 2007, 28 Suppl A:S64–9.Google Scholar
Doheny, HC, Houlihan, DD, Ravikumar, N, et al. Human chorionic gonadotrophin relaxation of human pregnant myometrium and activation of the BKCa channel. J Clin Endocrinol Metab. 2003, 88(9):43104315.Google Scholar
Karolczak-Bayatti, M, Loughney, AD, Robson, SC, et al. Epigenetic modulation of the protein kinase A RIIalpha (PRKAR2A) gene by histone deacetylases 1 and 2 in human smooth muscle cells. J Cell Mol Med. 2011, 15(1):94108.Google Scholar
Anamthathmakula, P, Kyathanahalli, C, Ingles, J, et al. Estrogen receptor alpha isoform ERdelta7 in myometrium modulates uterine quiescence during pregnancy. EBioMedicine. 2019, 39:520530.Google Scholar
Jones, SA, and Challis, JR. Effects of corticotropin-releasing hormone and adrenocorticotropin on prostaglandin output by human placenta and fetal membranes. Gynecol Obstet Invest. 1990, 29(3):165168.Google Scholar
Mesiano, S, Chan, EC, Fitter, JT, et al. Progesterone withdrawal and estrogen activation in human parturition are coordinated by progesterone receptor A expression in the myometrium. J Clin Endocrinol Metab. 2002, 87(6):29242930.Google Scholar
Nadeem, L, Balendran, R, Dorogin, A, et al. Pro-inflammatory signals induce 20alpha-HSD expression in myometrial cells: A key mechanism for local progesterone withdrawal. J Cell Mol Med. 2021.Google Scholar
Brizzi, P, Tonolo, G, Esposito, F, et al. Lipoprotein metabolism during normal pregnancy. Am J Obstet Gynecol. 1999, 181(2):430434.Google Scholar
Angelin, B, Olivecrona, H, Reihner, E, et al. Hepatic cholesterol metabolism in estrogen-treated men. Gastroenterology. 1992, 103(5):16571663.Google Scholar
Faggiano, A, Pivonello, R, Melis, D, et al. Evaluation of circulating levels and renal clearance of natural amino acids in patients with Cushing’s disease. J Endocrinol Invest. 2002, 25(2):142151.Google Scholar
Bridges, RS, Robertson, MC, Shiu, RP, et al. Endocrine communication between conceptus and mother: placental lactogen stimulation of maternal behavior. Neuroendocrinology. 1996, 64(1):5764.Google Scholar
Bridges, RS. Neuroendocrine regulation of maternal behavior. Front Neuroendocrinol. 2015, 36:178196.Google Scholar
Schiller, CE, Meltzer-Brody, S, and Rubinow, DR. The role of reproductive hormones in postpartum depression. CNS Spectr. 2015, 20(1):4859.Google Scholar
Macias, H, and Hinck, L. Mammary gland development. Wiley Interdiscip Rev Dev Biol. 2012, 1(4):533557.Google Scholar

References

Ivell, R, Ludwig, M, Tribe, RM, et al. Oxytocin. In Skinner, M. K. (Ed.), Encyclopedia of Reproduction. vol. 2, 2018; 597606. Academic Press: Elsevier.Google Scholar
Arrowsmith, S, and Wray, S. Oxytocin: Its mechanism of action and receptor signalling in the myometrium. J Neuroendocrinol. 2014, 26: 356369.Google Scholar
Borrow, AP, and Cameron, NM. The role of oxytocin in mating and pregnancy. Horm Behav. 2012, 61 :266276.Google Scholar
Giraldi, A, Marson, L, Nappi, R, et al. Physiology of female sexual function: Animal models. J Sexual Med. 2004, 1: 237253.Google Scholar
Nishimori, K, Young, LJ, Guo, Q et al. Oxytocin is required for nursing but is not essential for parturition or reproductive behavior. Proc Natl Acad Sci USA. 1996, 93: 1169911704.Google Scholar
Salonia, A, Nappi, RE, Pontillo, M, et al. Menstrual cycle-related changes in plasma oxytocin are relevant to normal sexual function in healthy women. Horm Behav. 2005, 47: 164169.Google Scholar
Veening, JG, de Jong, TR, Waldinger, MD, et al. The role of oxytocin in male and female reproductive behavior. Eur J Pharmacol. 2015, 753: 209228.Google Scholar
Carmichael, MS, Warburton, VL, Dixen, J, et al. Relationships among cardiovascular, muscular, and oxytocin responses during human sexual activity. Arch Sexual Behav. 1994, 23: 5979.Google Scholar
Einspanier, A, Ivell, R, and Hodges, JK. Oxytocin: A follicular luteinisation factor in the marmoset monkey. Adv Exp Med Biol. 1995, 395: 517522.Google Scholar
Furuya, K, Mizumoto, Y, Makimura, N, et al. A novel biological aspect of ovarian oxytocin: Gene expression of oxytocin and oxytocin receptor in cumulus/luteal cells and the effect of oxytocin on embryogenesis in fertilized oocytes. Adv Exp Med Biol. 1995, 395: 523528.Google Scholar
Fuchs, AR, Romero, R, Keefe, D, et al. Oxytocin secretion and human parturition: pulse frequency and duration increase during spontaneous labor in women. Am J Obstet Gynecol. 1991, 165: 15151523.Google Scholar
Wathes, DC, Borwick, SC, Timmons, PM, et al. Oxytocin receptor expression in human term and preterm gestational tissues prior to and following the onset of labour. J Endocrinol. 1999, 161: 143151.Google Scholar
Kimura, T, Takemura, M, Nomura, S, et al. Expression of oxytocin receptor in human pregnant myometrium. Endocrinology. 1996, 137: 780785.Google Scholar
Soloff, MS, Alexandrova, M, and Fernstrom, MJ. Oxytocin receptors: Triggers for parturition and lactation? Science. 1979, 204: 13131315.Google Scholar
Fuchs, AR, Fuchs, F, Husslein, P, et al. Oxytocin receptors in the human uterus during pregnancy and parturition. Am J Obstet Gynecol. 1984, 150: 734741.Google Scholar
Pieber, D, Allport, VC, Hills, F, et al. Interactions between progesterone receptor isoforms in myometrial cells in human labour. Mol Hum Reprod 2001, 7: 875879.Google Scholar
Mesiano, S, Chan, EC, Fitter, JT, et al. Progesterone withdrawal and estrogen activation in human parturition are coordinated by progesterone receptor A expression in the myometrium. J Clin Endocrinol Metab. 2002, 87: 29242930.Google Scholar
McNeilly, AS, Robinson, IC, Houston, MJ, et al. Release of oxytocin and prolactin in response to suckling. Br Med J. 1983, 286: 257259.Google Scholar
Uvnäs-Moberg, K, Widström, AM, Werner, S, et al. Oxytocin and prolactin levels in breast-feeding women. Correlation with milk yield and duration of breast-feeding. Acta Obstet Gynecol Scand. 1990, 69: 301306.Google Scholar
Fuchs, AR, Ivell, R, Ganz, N, et al. Secretion of oxytocin in pregnant and parturient cows: Corpus luteum may contribute to plasma oxytocin at term. Biol. Reprod. 2001, 65: 11351141.Google Scholar
Christensson, K, Nilsson, BA, Stock, S, et al. Effect of nipple stimulation on uterine activity and on plasma levels of oxytocin in full term, healthy, pregnant women. Acta Obstet Gynecol Scand. 1989, 68: 205210.Google Scholar
Jonas, W, Johansson, LM, Nissen, E, et al. Effects of intrapartum oxytocin administration and epidural analgesia on the concentration of plasma oxytocin and prolactin, in response to suckling during the second day postpartum. Breastfeeding Med. 2009, 4: 7182.Google Scholar
Nissen, E, Uvnäs-Moberg, K, Svensson, K, et al. Different patterns of oxytocin, prolactin but not cortisol release during breastfeeding in women delivered by caesarean section or by the vaginal route. Early Hum Devel. 1996, 45: 103118.Google Scholar
Yokoyama, Y, Ueda, T, Irahara, M, et al. Releases of oxytocin and prolactin during breast massage and suckling in puerperal women. Eur J Obstet Gynecol Reprod Biol. 1994, 53: 1720.Google Scholar
Uvnas Moberg, K, Ekstrom-Bergstrom, A, Buckley, S, et al. Maternal plasma levels of oxytocin during breastfeeding – A systematic review. PLoS ONE 2020, 15: e0235806.Google Scholar
Bell, AF, Erickson, EN, and Carter, CS: Beyond labor: The role of natural and synthetic oxytocin in the transition to motherhood. J Midwifery Womens Health. 2014, 59: 3542.Google Scholar
Odent, MR. Synthetic oxytocin and breastfeeding: Reasons for testing an hypothesis. Med Hypotheses. 2013, 81: 889891.Google Scholar
Williams, GL, Gazal, OS, Leshin, LS, et al. Physiological regulation of maternal behavior in heifers: roles of genital stimulation, intracerebral oxytocin release, and ovarian steroids. Biol Reprod. 2001, 65: 295300.Google Scholar
Carter, CS, Altemus, M, and Chrousos, GP. Neuroendocrine and emotional changes in the post-partum period. Prog Brain Res. 2001, 133: 241249.Google Scholar
Boccia, ML, Goursaud, AP, Bachevalier, J, et al. Peripherally administered non-peptide oxytocin antagonist, L368,899, accumulates in limbic brain areas: a new pharmacological tool for the study of social motivation in non-human primates. Horm Behav. 2007, 52: 344351.Google Scholar
Gordon, I, Zagoory-Sharon, O, Leckman, JF, et al. Oxytocin and the development of parenting in humans. Biol Psychiatry. 2010, 68: 377382.Google Scholar
Budden, A, Chen, LJ, and Henry, A. High-dose versus low-dose oxytocin infusion regimens for induction of labour at term. Cochrane Database Syst Rev. 2014, Cd009701.Google Scholar
Saccone, G, Ciardulli, A, Baxter, JK, et al. Discontinuing oxytocin infusion in the active phase of labor: A systematic review and meta-analysis. Obstet Gynec. 2017, 130: 10901096.Google Scholar
Tribe, RM, Crawshaw, SE, Seed, P, et al. Pulsatile versus continuous administration of oxytocin for induction and augmentation of labor: Two randomized controlled trials. Am J Obstet Gynecol. 2012, 206(230):e231–238.Google Scholar
ACOG Practice Bulletin No. 107: Induction of labor. Obstet Gynecol. 2009, 114: 386397.CrossRefGoogle Scholar
Smyth, RM, Markham, C, and Dowswell, T. Amniotomy for shortening spontaneous labour. Cochrane Database Syst Rev. 2013, Cd006167.Google Scholar
Grobman, WA, Rice, MM, Reddy, UM, et al. Labor induction versus expectant management in low-risk nulliparous women. N Engl J Med. 2018, 379: 513523.Google Scholar
Winterfeld, U, Meyer, Y, Panchaud, A, et al. Management of deficient lactation in Switzerland and Canada: A survey of midwives’ current practices. Breastfeeding Med. 2012, 7: 317318.Google Scholar
Cazorla-Ortiz, G, Obregón-Guitérrez, N, Rozas-Garcia, MR, et al. Methods and success factors of induced lactation: A scoping review. J Hum Lact. 2020, 36: 739749.Google Scholar
Mangesi, L, and Zakarija-Grkovic, I. Treatments for breast engorgement during lactation. Cochrane Database Syst Rev. 2016, Cd006946.Google Scholar
Fewtrell, MS, Loh, KL, Blake, A, et al. Randomised, double blind trial of oxytocin nasal spray in mothers expressing breast milk for preterm infants. Arch Dis Child Fetal Neonatal Ed. 2006, 91: F169–174.Google Scholar
Practice Bulletin No. 183: Postpartum Hemorrhage. Obstet Gynecol. 2017, 130: e168e186.Google Scholar
Salati, JA, Leathersich, SJ, Williams, MJ, et al. Prophylactic oxytocin for the third stage of labour to prevent postpartum haemorrhage. Cochrane Database Syst Rev. 2019, Cd001808.Google Scholar
Westhoff, G, Cotter, AM, and Tolosa, JE. Prophylactic oxytocin for the third stage of labour to prevent postpartum haemorrhage. Cochrane Database Syst Rev. 2013, Cd001808.Google Scholar
Durocher, J, Dzuba, IG, Carroli, G, et al. Does route matter? Impact of route of oxytocin administration on postpartum bleeding: A double-blind, randomized controlled trial. PLoS ONE. 2019, 14: e0222981.Google Scholar
Windle, RJ, Shanks, N, Lightman, SL, et al. Central oxytocin administration reduces stress-induced corticosterone release and anxiety behavior in rats. Endocrinology. 1997, 138: 28292834.Google Scholar
Petersson, M, Wiberg, U, Lundeberg, T, et al. Oxytocin decreases carrageenan induced inflammation in rats. Peptides. 2001, 22: 14791484.Google Scholar
Neumann, ID. Brain oxytocin: A key regulator of emotional and social behaviours in both females and males. J Neuroendocrinol. 2008, 20: 858865.Google Scholar
Churchland, PS, and Winkielman, P. Modulating social behavior with oxytocin: how does it work? What does it mean? Horm Behav. 2012, 61: 392399.Google Scholar
Cyranowski, JM, Hofkens, TL, Frank, E, et al. Evidence of dysregulated peripheral oxytocin release among depressed women. Psychosom Med. 2008, 70: 967975.Google Scholar
Leng, G, and Ludwig, M. Intranasal oxytocin: Myths and delusions. Biol Psychiatry. 2016, 79: 243250.Google Scholar
Saxbe, D, Khaled, M, Horton, KT, et al. Maternal prenatal plasma oxytocin is positively associated with prenatal psychological symptoms, but method of immunoassay extraction may affect results. Biol Psychol 2019. 147: 107718.Google Scholar
Bosch, OJ, Meddle, SL, Beiderbeck, DI, et al. Brain oxytocin correlates with maternal aggression: Link to anxiety. J Neurosci. 2005, 25: 68076815.Google Scholar
Craciunas, L, Kollmann, M, Tsampras, N, et al. Oxytocin antagonists for assisted reproduction. Cochrane Database Syst Rev. 2016, Cd012375.Google Scholar
Moraloglu, O, Tonguc, E, Var, T, et al. Treatment with oxytocin antagonists before embryo transfer may increase implantation rates after IVF. Reprod Biomed Online. 2010, 21: 338343.Google Scholar
Kam, KY, and Lamont, RF. Developments in the pharmacotherapeutic management of spontaneous preterm labor. Expert Opin Pharmacother. 2008, 9: 11531168.Google Scholar
van Vliet, EOG, Nijman, TAJ, Schuit, E, et al. Nifedipine versus atosiban for threatened preterm birth (APOSTEL III): A multicentre, randomised controlled trial. Lancet. 2016, 387: 21172124.Google Scholar
Thornton, S, Goodwin, TM, Greisen, G, et al. The effect of barusiban, a selective oxytocin antagonist, in threatened preterm labor at late gestational age: A randomized, double-blind, placebo-controlled trial. Am J Obstet Gynecol. 2009, 200(627): e621–610.Google Scholar
Thornton, S, Miller, H, Valenzuela, G, et al. Treatment of spontaneous preterm labour with retosiban: A phase 2 proof-of-concept study. Br J Clin Pharmacol. 2015, 80: 740749.Google Scholar
Ogawa, S, Kudo, S, Kitsunai, Y, et al. Increase in oxytocin secretion at ejaculation in male. Clin Endocrinol. 1980, 13: 9597.Google Scholar
Carmichael, MS, Humbert, R, Dixen, J, et al. Plasma oxytocin increases in the human sexual response. J Clin Endocrinol Metab. 1987, 64: 2731.Google Scholar
Melis, MR, Argiolas, A, andGessa, GL. Oxytocin-induced penile erection and yawning: site of action in the brain. Brain Res. 1986, 398: 259265.Google Scholar
Filippi, S, Vannelli, GB, Granchi, S, et al. Identification, localization and functional activity of oxytocin receptors in epididymis. Mol Cell Endocrinol. 2002, 193: 89100.Google Scholar
Ivell, R, Balvers, M, Rust, W, et al. Oxytocin and male reproductive function. Adv Exp Med Biol. 1997, 424: 253264.Google Scholar
Thackare, H, Nicholson, HD, and Whittington, K. Oxytocin - Its role in male reproduction and new potential therapeutic uses. Hum Reprod Update. 2006, 12: 437448.Google Scholar
Neumann, ID, Wigger, A, Torner, L, et al. Brain oxytocin inhibits basal and stress-induced activity of the hypothalamo-pituitary-adrenal axis in male and female rats: Partial action within the paraventricular nucleus. J Neuroendocrinol. 2000, 12: 235243.Google Scholar
Goodin, BR, Anderson, AJB, Freeman, EL, et al. Intranasal oxytocin administration is associated with enhanced endogenous pain inhibition and reduced negative mood states. Clin J Pain. 2015, 31: 757767.Google Scholar
Boll, S, Almeida de Minas, AC, Raftogianni, A, et al. Oxytocin and pain perception: From animal models to human research. Neuroscience. 2018, 387: 149161.Google Scholar
Tracy, LM, Georgiou-Karistianis, N, Gibson, SJ, et al. Oxytocin and the modulation of pain experience: Implications for chronic pain management. Neurosci Biobehav Rev. 2015, 55: 5367.Google Scholar
Onaka, T, and Takayanagi, Y. Role of oxytocin in the control of stress and food intake. J Neuroendocrinol. 2019, e12700.Google Scholar

References

Pierce, JG, and Parsons, TF. Glycoprotein hormones: Structure and function. Annu Rev Biochem. 1981, 50: 465495.Google Scholar
Stockell Hartree, A, and Renwick, AG. Molecular structures of glycoprotein hormones and functions of their carbohydrate components. Biochem J. 1992, 287(Pt 3): 665679.Google Scholar
Godine, JE, Chin, WW, and Habener, JF. Alpha Subunit of rat pituitary glycoprotein hormones. Primary structure of the precursor determined from the nucleotide sequence of cloned cDNAs. J Biol Chem. 1982, 257, 83688371,Google Scholar
Golos, TG, Durning, M, and Fisher, JM. Molecular cloning of the rhesus glycoprotein hormone alpha-subunit gene. DNA Cell Biol. 1991, 10, 367380.Google Scholar
McGregor, WG, Kuhn, RW, and Jaffe, RB. Biologically active chorionic gonadotropin: Synthesis by the human fetus. Science. 1983, 220, 306308.Google Scholar
Miller-Lindholm, AK, LaBenz, CJ, Ramey, J, et al. Human chorionic gonadotropin-beta gene expression in first trimester placenta. Endocrinology. 1997, 138, 54595465.Google Scholar
Morrish, DW, Manickavel, V, Jewell, LD, et al. Immunolocalization of alpha and beta chains of human chorionic gonadotropin, placental lactogen and pregnancy-specific beta-glycoprotein in term placenta by a touch preparation method. Histochemistry. 1987, 88, 5760.Google Scholar
Hoshina, M, Ashitake, Y, and Tojo, S. Immunohistochemical interaction on antisera to HCG and its subunits with chorionic tissue of early gestation. Endocrinol Jpn. 1979, 26, 175184.Google Scholar
Maruo, T, Ladines-Llave, CA, Matsuo, H, et al. A novel change in cytologic localization of human chorionic gonadotropin and human placental lactogen in first-trimester placenta in the course of gestation. Am J Obstet Gynecol. 1992, 167, 217222.Google Scholar
Beck, T, Schweikhart, G, and Stolz, E. Immunohistochemical location of HPL, SP1 and beta-HCG in normal placentas of varying gestational age. Arch Gynecol. 1986, 239, 6374.Google Scholar
Hoshina, M, Boothby, M, and Boime, I. Cytological localization of chorionic gonadotropin alpha and placental lactogen mRNAs during development of the human placenta. J Cell Biol. 1982, 93, 190198Google Scholar
Cole, LA. Immunoassay of human chorionic gonadotropin, its free subunits, and metabolites. Clin Chem. 1997, 43, 22332243.Google Scholar
Wolfahrt, S, Kleine, B, and Rossmanith, WG. Detection of gonadotrophin releasing hormone and its receptor mRNA in human placental trophoblasts using in-situ reverse transcription-polymerase chain reaction. Mol Hum Reprod. 1998, 4, 9991006.Google Scholar
Petraglia, F, Vaughan, J, and Vale, W. Inhibin and activin modulate the release of gonadotropin-releasing hormone, human chorionic gonadotropin, and progesterone from cultured human placental cells. Proc Natl Acad Sci USA. 1989, 86, 51145117.Google Scholar
Wehmann, RE, and Nisula, BC. Renal clearance rates of the subunits of human chorionic gonadotropin in man. J Clin Endocrinol Metab. 1980, 50, 674679.Google Scholar
Zirkin, BR, and Papadopoulos, V. Leydig cells: Formation, function, and regulation. Biol Reprod. 2018, 99, 101111.Google Scholar
Clements, JA, Reyes, FI, Winter, JS, et al. Studies on human sexual development. III. Fetal pituitary and serum, and amniotic fluid concentrations of LH, CG, and FSH. J Clin Endocrinol Metab. 1976, 42, 919.Google Scholar
Molsberry, RL, Carr, BR, Mendelson, CR, et al. Human chorionic gonadotropin binding to human fetal testes as a function of gestational age. J Clin Endocrinol Metab. 1982, 55, 791794.Google Scholar
Tapanainen, J, Kellokumpu-Lehtinen, P, Pelliniemi, L, et al. Age-related changes in endogenous steroids of human fetal testis during early and midpregnancy. J Clin Endocrinol Metab. 1981, 52, 98102.Google Scholar
Leinonen, PJ, and Jaffe, RB. Leydig cell desensitization by human chorionic gonadotropin does not occur in the human fetal testis. J Clin Endocrinol Metab. 1985, 61, 234238.Google Scholar
Hershman, JM. Human chorionic gonadotropin and the thyroid: Hyperemesis gravidarum and trophoblastic tumors. Thyroid. 1999, 9, 653657.Google Scholar
Kraiem, Z, Sadeh, O, Blithe, DL, et al. Human chorionic gonadotropin stimulates thyroid hormone secretion, iodide uptake, organification, and adenosine 3’,5’-monophosphate formation in cultured human thyrocytes. J Clin Endocrinol Metab. 1994, 79, 595599.Google Scholar
Tomer, Y, Huber, GK, and Davies, TF. Human chorionic gonadotropin (hCG) interacts directly with recombinant human TSH receptors. J Clin Endocrinol Metab. 1992, 74, 14771479.Google Scholar
Fitzpatrick, DL, and Russell, MA. Diagnosis and management of thyroid disease in pregnancy. Obstet Gynecol Clin North Am. 2010, 37, 173193.Google Scholar
Nathan, N, and Sullivan, SD. Thyroid disorders during pregnancy. Endocrinol Metab Clin North Am. 2014, 43, 573597.Google Scholar
Oppenheimer, JH, and Schwartz, HL. Molecular basis of thyroid hormone-dependent brain development. Endocr Rev, 1997, 18, 462475.Google Scholar
Noten, AM, Loomans, EM, Vrijkotte, TG, et al. Maternal hypothyroxinaemia in early pregnancy and school performance in 5-year-old offspring. Eur J Endocrinol. 2015, 173, 563571.Google Scholar
Nelson, SM, Haig, C, McConnachie, A, et al. Maternal thyroid function and child educational attainment: Prospective cohort study. BMJ. 2018, 360, k452.Google Scholar
Remaud, S, Gothie, JD, Morvan-Dubois, G, et al. Thyroid hormone signaling and adult neurogenesis in mammals. Front Endocrinol (Lausanne). 2014, 5, 62.Google Scholar
Kurtzman, JT, Wilson, H, and Rao, CV. A proposed role for hCG in clinical obstetrics. Semin Reprod Med. 2001, 19, 6368.Google Scholar
Kane, N, Kelly, R, Saunders, PT, et al. Proliferation of uterine natural killer cells is induced by human chorionic gonadotropin and mediated via the mannose receptor. Endocrinology. 2009, 150, 28822888.Google Scholar
Han, T. Inhibitory effect of human chorionic gonadotrophin on lymphocyte blastogenic response to mitogen, antigen and allogeneic cells. Clin Exp Immunol. 1974, 18, 529535.Google Scholar
Braunstein, GD. The long gestation of the modern home pregnancy test. Clin Chem. 2014, 60, 1821.Google Scholar
Wilcox, AJ, Baird, DD, Dunson, D, et al. Natural limits of pregnancy testing in relation to the expected menstrual period. JAMA. 2001, 286, 17591761.Google Scholar
Braunstein, GD. False-positive serum human chorionic gonadotropin results: Causes, characteristics, and recognition. Am J Obstet Gynecol. 2002, 187, 217224.Google Scholar
Montagnana, M, Trenti, T, Aloe, R, et al. Human chorionic gonadotropin in pregnancy diagnostics. Clin Chim Acta. 2011, 412, 15151520.Google Scholar
Grenache, DG. Variable accuracy of home pregnancy tests: Truth in advertising? Clin Chem Lab Med. 2015, 53, 339341.Google Scholar
Johnson, S, Cushion, M, Bond, S, et al. Comparison of analytical sensitivity and women’s interpretation of home pregnancy tests. Clin Chem Lab Med. 2015, 53, 391402.Google Scholar
Cole, LA. The utility of six over-the-counter (home) pregnancy tests. Clin Chem Lab Med. 2011, 49, 13171322.Google Scholar
Banerjee, S, Smallwood, A, Chambers, AE, et al. A link between high serum levels of human chorionic gonadotrophin and chorionic expression of its mature functional receptor (LHCGR) in Down’s syndrome pregnancies. Reprod Biol Endocrinol. 2005, 3, 25.Google Scholar
Stenman, UH, Alfthan, H, and Hotakainen, K. Human chorionic gonadotropin in cancer. Clin Biochem. 2004, 37, 549561.Google Scholar
Bedi, DG, Moeller, D, Fagan, CJ, et al. Chronic ectopic pregnancy. A comparison with acute ectopic pregnancy. Eur J Radiol. 1987, 7, 4648.Google Scholar
Brennan, DF, Kwatra, S, Kelly, M, et al. Chronic ectopic pregnancy – Two cases of acute rupture despite negative beta hCG. J Emerg Med. 2000, 19, 249254.Google Scholar
Smith, HO, Kohorn, E, and Cole, LA. Choriocarcinoma and gestational trophoblastic disease. Obstet Gynecol Clin North Am. 2005, 32, 661684.Google Scholar
Barnhart, K, Mennuti, MT, Benjamin, I, et al. Prompt diagnosis of ectopic pregnancy in an emergency department setting. Obstet Gynecol. 1994, 84, 10101015.Google Scholar
Barnhart, K, Sammel, MD, Chung, K, et al. Decline of serum human chorionic gonadotropin and spontaneous complete abortion: Defining the normal curve. Obstet Gynecol. 2004, 104, 975981.Google Scholar
Seeber, BE, Sammel, MD, Guo, W, et al. Application of redefined human chorionic gonadotropin curves for the diagnosis of women at risk for ectopic pregnancy. Fertil Steril. 2006, 86, 454459.Google Scholar
Chung, K, Sammel, MD, Coutifaris, C, et al. Defining the rise of serum HCG in viable pregnancies achieved through use of IVF. Hum Reprod. 2006, 21, 823828.Google Scholar
Silva, C, Sammel, MD, Zhou, L, et al. Human chorionic gonadotropin profile for women with ectopic pregnancy. Obstet Gynecol. 2006, 107, 605610.Google Scholar
Zee, J, Sammel, MD, Chung, K, et al. Ectopic pregnancy prediction in women with a pregnancy of unknown location: Data beyond 48 h are necessary. Hum Reprod. 2014, 29, 441447.Google Scholar

References

Abbassi-Ghanavati, M, Greer, LG, and Cunningham, FG. Pregnancy and laboratory studies: A reference table for clinicians. Obstet Gynecol. 2009, 114(6):13261331.Google Scholar
Devroey, P, Camus, M, Palermo, G, et al. Placental production of estradiol and progesterone after oocyte donation in patients with primary ovarian failure. Am J Obstet Gynecol. 1990, 162(1): 6670.Google Scholar
Berkane, N, Liere, P, Oudinet, JP, et al. From pregnancy to preeclampsia: A key role for estrogens. Endocr Rev. 2017, 38:123144.Google Scholar
Osawa, N Y. Purification of human placental aromatase cytochrome P-450 with monoclonal antibody and its characterization. Biochemistry. 1991, 30(12):30033010.Google Scholar
Pepe, GJ, and Albrecht, ED. Actions of placental and fetal adrenal steroid hormones in primate pregnancy. Endocr Rev. 1995, 16(5): 608648.Google Scholar
Siiteri, PK. The continuing saga of dehydroepiandrosterone (DHEA). J Clin Endocrinol Metab. 2005, 90(6):37953796.Google Scholar
Kragie, L. Aromatase in primate pregnancy: A review. Endocr Res. 2002, 28(3):121128.Google Scholar
Fournet-Dulguerov, N, MacLusky, NJ, Leranth, CZ, et al. Immunohistochemical localization of aromatase cytochrome P-450 and estradiol dehydrogenase in the syncytiotrophoblast of the human placenta. J Clin Endocrinol Metab. 1987, 65(4):757764.Google Scholar
Kumar, P, Kamat, A, and Mendelson, CR. Estrogen receptor alpha (Eralpha) mediates stimulatory effects of estrogen on aromatase (CYP19) gene expression in human placenta. Mol Endocrinol. 2009, 23(6):784793.Google Scholar
Calzada-Mendoza, CC, Sa´nchez, EC, Campos, RR, et al. Differential aromatase (CYP19) expression in human arteries from normal and neoplasic uterus: An immuno-histochemical and in situ hybridization study. Front Biosci. 2006, 11:389393.Google Scholar
Zbella, EA, Ilekis, J, Scommegna, A, et al. Competitive studies with dehydroepiandrosterone sulfate and 16 alpha- hydroxydehydroepiandrosterone sulfate in cultured human cho- riocarcinoma JEG-3 cells: Effect on estrone, 17 beta-estradiol, and estriol secretion. J Clin Endocrinol Metab. 1986, 63(3):751757.Google Scholar
Milewich, L, MacDonald, PC, and Carr, BR. Estrogen 16 alpha-hydroxylase activity in human fetal tissues. J Clin Endocrinol Metab. 1986, 63(2):404406.Google Scholar
Jobe, SO, Tyler, CT, and Magness, RR. Aberrant synthesis, metabolism, and plasma accumulation of circulating estrogens and estrogen metabolites in preeclampsia implications for vascular dysfunction. Hypertension. 2013, 61(2):480487.Google Scholar
Holinka, CF, Diczfalusy, E, and Coelingh Bennink, HJT. Estetrol: A unique steroid in human pregnancy. J Steroid Biochem Mol Biol. 2008, 110(1–2):138143.Google Scholar
Pluchino, N, Santoro, AN, Casarosa, E, et al. Effect of estetrol administration on brain and serum allopregnanolone in intact and ovariectomized rats. J Steroid Biochem Mol Biol. 2014, 143: 285290.Google Scholar
Jobe, SO, Ramadoss, J, Koch, JM, et al. Estradiol-17beta and its cytochrome P450- and catechol-O-methyltransferase-derived metabolites stimulate proliferation in uterine artery endothelial cells: Role of estrogen receptor-alpha versus estrogen receptor-beta. Hypertension. 2010, 55(4): 10051011.Google Scholar
Aitio, A. UDPglucuronosyltransferase of the human placenta. Biochem Pharmacol. 1974, 23(15):22032205.Google Scholar
Gamage, N, Barnett, A, Hempel, N, et al. Human sulfotransferases and their role in chemical metabolism. Toxicol Sci. 2006, 90(1):522.Google Scholar
Falany, CN, Wheeler, J, Oh, TS, et al. Steroid sulfation by expressed human cytosolic sulfotransferases. J Steroid Biochem Mol Biol. 1994, 48(4):369375.Google Scholar
Tong, MH, Jiang, H, Liu, P, et al. Spontaneous fetal loss caused by placental thrombosis in estrogen sulfotransferase-deficient mice. Nat Med. 2005, 11(2):153159.Google Scholar
Pepe, GJ, Burch, MG,and Albrecht, E. Estroge regulates 11ß-hydroxysteroid dehydrogenase-1 and -2 localization in placental syncytiotrophoblast in the second half of primate pregnancy. Endocrinology. 2001, 142.44964503.Google Scholar
Hu, W, Weng, X, Dong, M, et al. Alteration in methylation level at 11ß-hydroxysteroid dehydrogenase type 2 gene promoter in infants born to preeclamptic women. BMC Genetics. 2014, 15:96.Google Scholar
Dao, B, Vanageg, MA, Bardin, CW, et al. Anti-implantation activity of antiestrogens and mifepristone. Contraception. 1996, 54: 253258.Google Scholar
Bukovsky, A, Cekanova, M, Caudle, MR, et al. Expression and localization of estrogen receptor-alpha protein in normal and abnormal term placentae and stimulation of trophoblast differentiation by estradiol. Reprod Biol Endocrinol. 2003, 1:13.Google Scholar
Bukovsky, A, Caudle, MR, Cekanova, M, et al. Placental expression of estrogen receptor beta and its hormone binding variant – Comparison with estrogen receptor alpha and a role for estrogen receptors in asymmetric division and differentiation of estrogen-dependent cells. Reprod Biol Endocrinol. 2003, 1:36.Google Scholar
Pijnenborg, R, Vercruysse, L, and Hanssens, M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta. 2006, 27(9–10):939958.Google Scholar
Liu, LX, Lu, H, Luo, Y, et al. Stabilization of vascular endothelial growth factor mRNA by hypoxia-inducible factor 1. Biochem Biophys Res Commun. 2002, 291(4):908914.Google Scholar
Losordo, DW, and Isner, JM. Estrogen and angiogenesis: A review. Arterioscler Thromb Vasc Biol. 2001, 21(1):612.Google Scholar
Ce, ZM, Bucak, K, Chu, S, et al. 17Beta-estradiol up-regulates vascular endothelial growth factor receptor-2 expression in human myometrial microvascular endothelial cells: Role of estrogen receptor-alpha and -beta. J Clin Endocrinol Metab. 2002, 87(9):43414349.Google Scholar
Fotsis, T, Zhang, Y, Pepper, MS, et al. The endogenous oestrogen metabolite 2-methoxyoestradiol inhibits angiogenesis and suppresses tumour growth. Nature. 1994, 368(6468):237239.Google Scholar
Thaler, I, Manor, D, Itskovitz, J, et al. Changes in uterine blood flow during human pregnancy. Am J Obstet Gynecol. 1990, 162(1):121125.Google Scholar
Mabie, WC, DiSessa, TG, Crocker, LG, et al. A longitudinal study of cardiac output in normal human pregnancy. Am J Obstet Gynecol. 1994, 170(3):849856.Google Scholar
Dubey, RK, and Jackson, EK. Potential vascular actions of 2-methoxyestradiol. Trends Endocrinol Metab. 2009,20(8):374379.Google Scholar
Rupnow, HL, Phernetton, TM, Shaw, CE, et al. Endothelial vasodilator production by uterine and systemic arteries. VII. Estrogen and progesterone effects on eNOS. Am J Physiol Heart Circ Physiol. 2001, 280(4):H1699H1705.Google Scholar
Miller, SL, Jenkin, G, and Walker, DW. Effect of nitric oxide synthase inhibition on the uterine vasculature of the late-pregnant ewe. Am J Obstet Gynecol. 1999, 180(5):11381145.Google Scholar
Baggia, S, Albrecht, ED, and Pepe, GJ. Regulation of 11 beta-hydroxysteroid dehydrogenase activity in the baboon placenta by estrogen. Endocrinology. 1990, 126(5):27422748.Google Scholar
Almey, A, Milner, TA, and Brake, W. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm Behav. 2015, 74:125138.Google Scholar
Baud, O, Berkane, N. Hormonal changes associated with intra-uterine growth restriction: Impact on the developing brain and future neurodevelopment. Front Endocrinol. 2019, 10:179.Google Scholar
Hertig, A, Liere, P, Chabbert-Buffet, N, et al. Steroid profiling in preeclamptic women: Evidence for aromatase deficiency. Am J Obstet Gynecol. 2010, 203(5):477.e1–477.e9.Google Scholar
Perez-Sepulveda, A, Monteiro, LJ, Dobierzewska, A, et al. Placental aromatase is deficient in placental ischemia and preeclampsia. PLoS ONE. 2015, 10(10):e0139682.Google Scholar
Kumar, P, Luo, Y, Tudela, C, et al. The c-Myc-regulated microRNA-17~92 (miR-17~92) and miR- 106a~363 clusters target hCYP19A1 and hGCM1 to inhibit human trophoblast differentiation. Mol Cell Biol. 2013, 33(9): 17821796.Google Scholar
Ma, CX, Adjei, AA, Salavaggione, OE, et al. Human aromatase: Gene resequencing and functional genomics. Cancer Res. 2005, 65(23):1107111082.Google Scholar
Charles, SM, Julian, CG, Vargas, E, et al. Higher estrogen levels during pregnancy in Andean than European residents of high altitude suggest differences in aromatase activity. J Clin Endocrinol Metab. 2014, 99(8):29082916.Google Scholar
Berkane, N, Liere, P, Lefevre, G, et al. Abnormal steroidogenesis and aromatase activity and the risk of preeclampsia. Placenta. 2018, 69:4049.Google Scholar
Kanasaki, K, Palmsten, K, Sugimoto, H, et al. Deficiency in catechol-O-methyltransferase and 2-methoxyoestradiol is associated with pre-eclampsia. Nature. 2008, 453(7198):11171121.Google Scholar
Ishibashi, O, Ohkuchi, A, Ali, MM, et al. Hydroxysteroid (17-b) dehydrogenase 1 is dysregulated by miR-210 and miR-518c that are aberrantly expressed in preeclamptic placentas: a novel marker for predicting pre-eclampsia. Hypertension. 2012, 59(2):265273.Google Scholar
Ohkuchi, A, Ishibashi, O, Hirashima, C, et al. Plasma level of hydroxysteroid (17-b) dehydrogenase 1 in the second trimester is an independent risk factor for predicting preeclampsia after adjusting for the effects of mean blood pressure, bilateral notching and plasma level of soluble fms-like tyrosine kinase 1/placental growth factor ratio. Hypertens Res. 2012, 35(12):11521158.Google Scholar
Ludwikowski, B, Heger, S, Datz, N, et al. Aromatase deficiency: Rare cause of virilization. Eur J Pediatr Surg. 2013, 23(5):418422.Google Scholar
Maliqueo, M, Echiburú, B, and Crisosto, N. Sex steroids modulate uterine-placental vasculature: Implications for obstetrics and neonatal outcomes. Front Physiol. 2016, 7:152.Google Scholar
Salih, SM, Salama, SA, Fadl, AA, et al. Expression and cyclic variations of catechol-O-methyl transferase in human endometrial stroma. Fertil Steril. 2008, 90(3):789797.Google Scholar
Rabe, T, Hösch, R, and Runnebaum, B. Sulfatase deficiency in the human placenta: Clinical findings. Biol Res Pregnancy Perinatol. 1983, 4(3):95102.Google Scholar
Young, I, Renfree, M, Mesiano, S, et al. The comparative physiology of parturition in mammals: Hormones and parturition in mammals. 2011, 5:95116.Google Scholar
Renthal, NE, Williams, KC, Montalbano, AP, et al. Molecular regulation of parturition: A myometrial perspective. Cold Spring Harb Perspect Med. 2015, 5:a023069.Google Scholar
Weiss, G. Clinical review 118. Endocrinology of parturition. J Clin Endocrinol Metab. 2000, 85:44214425.Google Scholar

References

Callard, IP, Fileti, LA, Perez, LE, et al. Role of the corpus luteum and progesterone in the evolution of vertebrate viviparity. Am. Zool. 1992, 32:264275.Google Scholar
Hisaw, FL. Endocrine adaptations of the mammalian estrous cycle and gestation. In: Gorbman, A, ed. Comparative Endocrinology. 1959; 533552. New York.: John Wiley & Sons.Google Scholar
Wilson, SC, andSharp, PJ. The effects of progesterone on oviposition and ovulation in the domestic fowl (Gallus domesticus). Br Poult Sci. 1976, 17:163173.Google Scholar
Thornton, JW. Evolution of vertebrate steroid receptors from an ancestral estrogen receptor by ligand exploitation and serial genome expansions. Proc Natl Acad Sci USA. 2001, 98:56715676.Google Scholar
Corner, GW. The Hormones in Human Reproduction. London: Princeton University Press.Google Scholar
Allen, WM. Recollections of my life with progesterone. Gynecol Invest. 1974, 5:142182.Google Scholar
Corner, GW, Sr. The early history of progesterone. Gynecol Invest. 1974, 5:106112.Google Scholar
Medvei, VC. A History of Clinical Endocrinology. New York: The Parthenon Publishing Group.Google Scholar
Frobenius, W. Ludwig Fraenkel: ‘Spiritus rector’ of the early progesterone research. Eur J Obstet Gynecol Reprod Biol. 1999, 83:115119.Google Scholar
Allen, WM, and Wintersteiner, O. Crystalline progestin. Science. 1934, 80:190191.Google Scholar
Butenandt, A, andWestphal, U. Zur Isolierung und Characterisierung des Corpus-luteum-Hormons. Ber Chem Ges. 1934, 67:14401442.Google Scholar
Hartmann, M, and Wettstein, A. Ein krystallisiertes Hormon aus Corpus luteum. Helv Chim Acta. 1934, 17:878882.Google Scholar
Slotta, K, Ruschig, H, and Fels, E. Reindarstellung der Hormone aus dem Corpus luteum. Ber Chem Ges. 1934, 67:12701273.Google Scholar
Allen, WM, Butenandt, A, Corner, GW, et al. Nomenclature of corpus luteum hormone. Science. 1935, 82:153.Google Scholar
Davis, ME, and Wied, GL. 17-alpha-hydroxyprogesterone-caproate: A new substance with prolonged progestational activity: A comparison with chemically pure progesterone. J Clin Endocrinol Metabol. 1955, 15:923930.Google Scholar
Venning, EH, and Browne, JSL. Isolation of a water-soluble pregnandiol complex from human pregnancy urine. Proc Soc Exper Biol and Med. 1936, 34:729.Google Scholar
Venning, EH. Gravimetric method for the determination of sodium pregnandiol glucuronidate (an excretion product of progesterone). J Biol Chem. 1937, 119:473.Google Scholar
Wilson, KM, and Rochester, NY. Pregnancy complicated by ovarian and paraovarian tumors. Am J Ob/Gyn. 1937, 34:977986.Google Scholar
Csapo, AI, Pulkkinen, MO, Ruttner, B, et al. The significance of the human corpus luteum in pregnancy maintenance. I. Preliminary studies. Am J Obstet Gynecol. 1972, 112:10611067.Google Scholar
Csapo, A. The luteo-placental shift, the guardian of pre-natal life. Postgrad Med J. 1969, 45:5764.Google Scholar
O’Malley, BW. In vitro hormonal induction of a specific protein (avidin) in chick oviduct. Biochemistry. 1967, 6:25462551.Google Scholar
O’Malley, BW, and Korenman, SG. Studies on the mechanism of hormone induction of a specific protein. Immunological identity and kinetic studies of avidin synthesized in vitro by the chick oviduct. Life Sci. 1967, 6:19531959.Google Scholar
Milgrom, E, Atger, M, and Baulieu, EE. Progesterone in uterus and plasma. IV. Progesterone receptor(s) in guinea pig uterus cytosol. Steroids. 1970, 16:741754.Google Scholar
Smith, RG, Iramain, CA, Buttram, VC, Jr., et al. Purification of human uterine progesterone receptor. Nature. 1975, 253:271272.Google Scholar
Heikinheimo, O, Kontula, K, Croxatto, H, et al. Plasma concentrations and receptor binding of RU 486 and its metabolites in humans. J Steroid Biochem. 1987, 26:279284.Google Scholar
Gellersen, B, Brosens, IA, and Brosens, JJ. Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Sem Reprod Med. 2007, 25:445453.Google Scholar
Mao, G, Wang, J, Kang, Y, et al. Progesterone increases systemic and local uterine proportions of CD4+CD25+ Treg cells during midterm pregnancy in mice. Endocrinology. 2010, 151:54775488.Google Scholar
Renthal, NE, Chen, CC, Williams, KC, et al. miR-200 family and targets, ZEB1 and ZEB2, modulate uterine quiescence and contractility during pregnancy and labor. Proc Natl Acad Sci USA. 2010, 107:2082820833.Google Scholar
Siiteri, PK, Febres, F, Clemens, LE, et al. Progesterone and maintenance of pregnancy: is progesterone nature’s immunosuppressant? Ann NY Acad Sci. 1977, 286:384397.Google Scholar
Hardy, DB, Janowski, BA, Corey, DR, et al. Progesterone receptor plays a major antiinflammatory role in human myometrial cells by antagonism of nuclear factor-kappaB activation of cyclooxygenase 2 expression. Mol Endocrinol. 2006, 20:27242733.Google Scholar
Thomson, AJ, Telfer, JF, Young, A, et al. Leukocytes infiltrate the myometrium during human parturition: further evidence that labour is an inflammatory process. Hum Reproduct. 1999, 14:229236.Google Scholar
Osman, I, Young, A, Jordan, F, et al. Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term. J Soc Gynecol Invest. 2006, 13:97103.Google Scholar
Mesiano, S, Chan, EC, Fitter, JT, et al. Progesterone withdrawal and estrogen activation in human parturition are coordinated by progesterone receptor A expression in the myometrium. J Clin Endocrinol Metabol. 2002, 87:29242930.Google Scholar
Tan, H, Yi, L, Rote, NS, et al. Progesterone receptor-A and -B have opposite effects on proinflammatory gene expression in human myometrial cells: Implications for progesterone actions in human pregnancy and parturition. J Clin Endocrinol Metabol. 2012, 97:E719–730.Google Scholar
Merlino, AA, Welsh, TN, Tan, H, et al. Nuclear progesterone receptors in the human pregnancy myometrium: evidence that parturition involves functional progesterone withdrawal mediated by increased expression of progesterone receptor-A. J Clin Endocrinol Metabol. 2007, 92:19271933.Google Scholar
Chai, SY, Smith, R, Fitter, JT, et al. Increased progesterone receptor A expression in labouring human myometrium is associated with decreased promoter occupancy by the histone demethylase JARID1A. Mol Hum Reprod. 2014, 20:442453.Google Scholar
Amini, P, Michniuk, D, Kuo, K, et al. Human parturition involves phosphorylation of progesterone receptor-A at serine-345 in myometrial cells. Endocrinology. 2016, 157:44344445.Google Scholar
Condon, JC, Jeyasuria, P, Faust, JM, et al. A decline in the levels of progesterone receptor coactivators in the pregnant uterus at term may antagonize progesterone receptor function and contribute to the initiation of parturition. Proc Natl Acad Sci USA. 2003, 100:95189523.Google Scholar
Mahendroo, MS, Porter, A, Russell, DW, et al. The parturition defect in steroid 5alpha-reductase type 1 knockout mice is due to impaired cervical ripening. Mol Endocrinol. 1999, 13:981992.Google Scholar
Nadeem, L, Shynlova, O, Matysiak-Zablocki, E, et al. Molecular evidence of functional progesterone withdrawal in human myometrium. Nat Commun. 2016, 7:11565.Google Scholar
Nadeem, L, Shynlova, O, Mesiano, S, et al. Progesterone via its type-A receptor promotes myometrial gap junction coupling. Sci Rep. 2017, 7:13357.Google Scholar
Bengtsson, LP. The Effect of Progesterone on the Maintenance of Early Pregnancy. Paper presented at: Brook Lodge Symposium: Progesterone 1961; Augusta, MI.Google Scholar
Pinto, RM, Montuori, E, Lerner, U, et al. Effect of progesterone on the oxytocic action of estradiol-17-beta. Am J Obstet Gynecol. 1965, 91:10841089.Google Scholar
Keirse, MJ. Progestogen administration in pregnancy may prevent preterm delivery. Br J Obstet Gynaecol. 1990, 97:149154.Google Scholar
da Fonseca, EB, Bittar, RE, Carvalho, MH, et al. Prophylactic administration of progesterone by vaginal suppository to reduce the incidence of spontaneous preterm birth in women at increased risk: a randomized placebo-controlled double-blind study. Am J Obstet Gynecol. 2003, 188:419424.Google Scholar
Meis, PJ, Klebanoff, M, Thom, E, et al. Prevention of recurrent preterm delivery by 17 alpha-hydroxyprogesterone caproate. N Engl J Med. 2003, 348:23792385.Google Scholar
Norman, JE, Marlow, N, Messow, CM, et al. Vaginal progesterone prophylaxis for preterm birth (the OPPTIMUM study): A multicentre, randomised, double-blind trial. Lancet. 2016, 387:21062116.Google Scholar
Blackwell, SC, Gyamfi-Bannerman, C, Biggio, JR, Jr., et al. 17-OHPC to prevent recurrent preterm birth in singleton gestations (PROLONG Study): A multicenter, international. Randomized Double-Blind Trial. Am J Perinatol. 2020, 37:127136.Google Scholar
Weatherborn, M, and Mesiano, S. Rationale for current and future progestin-based therapies to prevent preterm birth. Best Prac Res Clin Obstet Gynaecol. 2018, 52:114125.Google Scholar

References

Spaanderman, M, and Staelens, A (2018). Plasma volume changes in pregnancy. In: C. Lees & W. Gyselaers (Eds.), Mater Hemodyn. Cambridge: Cambridge University Press. https://doi.org/10.1017/9781316661925.Google Scholar
Scholten, RR, Oyen, WJ, Van der Vlugt, MJ, et al. Impaired fetal growth and low plasma volume in adult life. Obstet Gynecol. 2011, 118(6):13141322. https://doi.org/10.1097/AOG.0b013e3182383781. PMID: 22105261.Google Scholar
Sanghavi, M, and Rutherford, JD. Cardiovascular physiology of pregnancy. Circulation. 2014, 130(12):10031008. https://doi.org/10.1161/CIRCULATIONAHA.114.009029. PMID: 25223771.Google Scholar
Ouzounian, JG, and Elkayam, U. Physiologic changes during normal pregnancy and delivery. Cardiol Clin. 2012, 30(3):317329. https://doi.org/10.1016/j.ccl.2012.05.004. Epub 2012. PMID: 22813360.Google Scholar
Chung, E, and Leinwand, LA. Pregnancy as a cardiac stress model. Cardiovasc Res. 2014, 101(4):561570. https://doi.org/10.1093/cvr/cvu013. Epub 201. PMID: 24448313; PMCID: PMC3941597.Google Scholar
Peeters, L. Cardiovascular and volume regulatory functions in pregnancy: An overview. In C. Lees & W. Gyselaers (Eds.), Maternal Hemodynamics. 2018; 13–23. Cambridge: Cambridge University Press. https://doi.org/10.1017/9781316661925.002.Google Scholar
Tal, R, Taylor, HS, Burney, RO, et al. Endocrinology of Pregnancy. In Feingold, KR, Anawalt, B, Boyce, A, Chrousos, G, de Herder, WW, Dungan, K, Grossman, A, Hershman, JM, Hofland, J, Kaltsas, G, Koch, C, Kopp, P, Korbonits, M, McLachlan, R, Morley, JE, New, M, Purnell, J, Singer, F, Stratakis, CA, Trence, DL, Wilson, DP, (Eds). 2000. South Dartmouth, MA: MDText.com, Inc..Google Scholar
Kodogo, V, Azibani, F, and Sliwa, K. Role of pregnancy hormones and hormonal interaction on the maternal cardiovascular system: A literature review. Clin Res Cardiol. 2019, 108(8):831846. https://doi.org/10.1007/s00392-019-01441-x. Epub 2019. PMID: 30806769.Google Scholar
Napso, T, Yong, HEJ, Lopez-Tello, J, et al. The role of placental hormones in mediating maternal adaptations to support pregnancy and lactation. Front Physiol. 2018, 9:1091. https://doi.org/10.3389/fphys.2018.01091. PMID: 30174608; PMCID: PMC6108594.Google Scholar
Das, A, Mantena, SR, Kannan, A, et al. De novo synthesis of estrogen in pregnant uterus is critical for stromal decidualization and angiogenesis. Proc Natl Acad Sci USA. 2009, 106(30):1254212547. https://doi.org/10.1073/pnas.0901647106.Google Scholar
Takahashi, K, Ohmichi, M, Yoshida, M, et al. Both estrogen and raloxifene cause G1 arrest of vascular smooth muscle cells. J Endocrinol. 2003, 178(2):319329. https://doi.org/10.1677/joe.0.1780319. PMID: 12904179.Google Scholar
Castardo-de-Paula, JC, de Campos, BH, Amorim, EDT, et al. Cardiovascular risk and the effect of nitric oxide synthase inhibition in female rats: The role of estrogen. Exp Gerontol. 2017, 97:3848. https://doi.org/10.1016/j.exger.2017.07.016. Epub 2017. PMID: 28757113.Google Scholar
Favre, J, Gao, J, Henry, J-P, et al. Endothelial estrogen receptor {alpha} plays an essential role in the coronary and myocardial protective effects of estradiol in ischemia/reperfusion. Arterioscler Thromb Vasc Biol. 2010, 30:25622567.Google Scholar
Fortini, F, Dalla Sega, FV, Caliceti, C, et al. Estrogen receptor β-dependent Notch1 activation protects vascular endothelium against tumor necrosis factor α (TNFα)-induced apoptosis. J Biol Chem. 2017, 292:1817818191.Google Scholar
Jobe, SO, Ramadoss, J, Koch, JM, et al. Estradiol-17beta and its cytochrome P450- and catechol-O-methyltransferase-derived metabolites stimulate proliferation in uterine artery endothelial cells: Role of estrogen receptor-alpha versus estrogen receptor-beta. Hypertension. 2010, 55(4):10051011. https://doi.org/10.1161/HYPERTENSIONAHA.109.146399. Epub 2010. PMID: 20212268; PMCID: PMC2876348.Google Scholar
Storment, JM, Meyer, M, and Osol, G. Estrogen augments the vasodilatory effects of vascular endothelial growth factor in the uterine circulation of the rat. Am J Obstet Gynecol. 2000, 183(2):449453. https://doi.org/10.1067/mob.2000.105910. PMID: 10942485.Google Scholar
van Eickels, M, Grohé, C, Cleutjens, JP, et al. 17beta-estradiol attenuates the development of pressure-overload hypertrophy. Circulation. 2001, 104(12):14191423. https://doi.org/10.1161/hc3601.095577. PMID: 11560859.Google Scholar
Bueno, OF, De Windt, LJ, Lim, HW, et al. The dual-specificity phosphatase MKP-1 limits the cardiac hypertrophic response in vitro and in vivo. Circ Res. 2001, 88(1):8896. https://doi.org/10.1161/01.res.88.1.88. PMID: 11139479.Google Scholar
Fliegner, D, Schubert, C, Penkalla, A, et al. Female sex and estrogen receptor-beta attenuate cardiac remodeling and apoptosis in pressure overload. Am J Physiol Regul Integr Comp Physiol. 2010, 298(6):R1597–606. https://doi.org/10.1152/ajpregu.00825. 2009. Epub 2010.Google Scholar
Liou, C-M, Yang, A-L, Kuo, C-H, et al. E ects of 17 Beta- estradiol on cardiac apoptosis in overiectomized rats. Cell Biochem Funct. 2010, 28:521528. https://doi.org/10.1016/j.numecd.2011.11.002.Google Scholar
Zhu, Y, Bian, Z, Lu, P, et al. Abnormal vascular function and hypertension in mice deficient in estrogen receptor beta. Science. 2002, 295(5554):505508. https://doi.org/10.1126/science.1065250. PMID: 11799247.Google Scholar
Eghbali, M, Deva, R, Alioua, A, et al. Molecular and functional signature of heart hypertrophy during pregnancy. Circ Res. 2005, 96(11):12081216. https://doi.org/10.1161/01.RES.0000170652.71414.16. Epub 2005. PMID: 15905459.Google Scholar
Karpanou, EA, Vyssoulis, GP, Georgoudi, DG, et al. Ambulatory blood pressure changes in the menstrual cycle of hypertensive women. Significance of plasma renin activity values. Am J Hypertens. 1993, 6(8):654659. https://doi.org/10.1093/ajh/6.8.654. PMID: 8217027.Google Scholar
Simoncini, T, Fu, XD, Caruso, A, et al. Drospirenone increases endothelial nitric oxide synthesis via a combined action on progesterone and mineralocorticoid receptors. Hum Reprod. 2007, 22(8):23252334. https://doi.org/10.1093/humrep/dem109. Epub 2007. PMID: 17545686.Google Scholar
Simoncini, T, Mannella, P, Fornari, L, et al. Differential signal transduction of progesterone and medroxyprogesterone acetate in human endothelial cells. Endocrinology. 2004, 145(12):57455756. https://doi.org/10.1210/en.2004-0510. Epub 2004. PMID: 15358673.Google Scholar
Zhang, L, Fishman, MC, and Huang, PL. Estrogen mediates the protective effects of pregnancy and chorionic gonadotropin in a mouse model of vascular injury. Arterioscler Thromb Vasc Biol. 1999, 19(9):20592065. https://doi.org/10.1161/01.atv.19.9.2059. PMID: 10479646.Google Scholar
Thomas, P, and Pang, Y. Protective actions of progesterone in the cardiovascular system: potential role of membrane progesterone receptors (mPRs) in mediating rapid effects. Steroids. 2013, 78(6):583588. https://doi.org/10.1016/j.steroids.2013.01.003. Epub 2013. PMID: 23357432.Google Scholar
Liu, LX, Rowe, GC, Yang, S, et al. PDK4 Inhibits Cardiac Pyruvate Oxidation in Late Pregnancy. Circ Res. 2017, 121(12):13701378. https://doi.org/10.1161/CIRCRESAHA.117.311456. Epub 2017. PMID: 28928113; PMCID: PMC5722682.Google Scholar
Chung, E, Yeung, F, and Leinwand, LA. Calcineurin activity is required for cardiac remodelling in pregnancy. Cardiovasc Res. 2013, 100(3):402410. https://doi.org/10.1093/cvr/cvt208. Epub 2013. PMID: 23985902; PMCID: PMC3826703.Google Scholar
Morrissy, S, Xu, B, Aguilar, D, et al. Inhibition of apoptosis by progesterone in cardiomyocytes. Aging Cell. 2010, 9(5):799809. https://doi.org/10.1111/j.1474-9726.2010.00619.x. PMID: 20726854; PMCID: PMC4133411.Google Scholar
Hsieh, DJ, Huang, CY, Pai, P, et al. Prolactin protects cardiomyocytes against intermittent hypoxia-induced cell damage by the modulation of signaling pathways related to cardiac hypertrophy and proliferation. Int J Cardiol. 2015, 181:255266. https://doi.org/10.1016/j.ijcard.2014.11.154. Epub 2014. PMID: 25531577.Google Scholar
Gonzalez, C, Rosas-Hernandez, H, Jurado-Manzano, B, et al. The prolactin family hormones regulate vascular tone through NO and prostacyclin production in isolated rat aortic rings. Acta Pharmacol Sin. 2015, 36(5):572586. https://doi.org/10.1038/aps.2014.159. Epub 2015. PMID: 25891087; PMCID: PMC4422941.Google Scholar
Zamani, P, and Greenberg, BH. Novel vasodilators in heart failure. Curr Heart Fail Rep. 2013, 10(1):111. https://doi.org/10.1007/s11897-012-0126-4. PMID: 23299783.Google Scholar
Kristiansson, P, and Wang, JX. Reproductive hormones and blood pressure during pregnancy. Hum Reprod. 2001, 16(1):1317. https://doi.org/10.1093/humrep/16.1.13. PMID: 11139529.Google Scholar
Smith, MC, Danielson, LA, Conrad, KP, et al. Influence of recombinant human relaxin on renal hemodynamics in healthy volunteers. J Am Soc Nephrol. 2006, 17(11):31923197. https://doi.org/10.1681/ASN.2005090950. Epub 2006. PMID: 17035617.Google Scholar
Leo, CH, Ng, HH, Marshall, SA, et al. Relaxin reduces endothelium-derived vasoconstriction in hypertension: Revealing new therapeutic insights. Br J Pharmacol. 2020, 177(1):217233. https://doi.org/10.1111/bph.14858. Epub 2019. PMID: 31479151; PMCID: PMC6976785.Google Scholar
Soma-Pillay, P, Nelson-Piercy, C, Tolppanen, H, et al. Physiological changes in pregnancy. Cardiovasc J Afr. 2016, 27(2):8994. https://doi.org/10.5830/CVJA-2016-021. PMID: 27213856; PMCID: PMC4928162.Google Scholar
Gennari-Moser, C, Khankin, EV, Schüller, S, et al. Regulation of placental growth by aldosterone and cortisol. Endocrinology. 2011, 152(1):263271. https://doi.org/10.1210/en.2010-0525. Epub 2010. PMID: 21068161.Google Scholar

Save book to Kindle

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

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

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

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

Available formats
×

Save book to Google Drive

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

Available formats
×