簡易檢索 / 詳目顯示

研究生: 邱惠芳
Chiu, Hui-Fang
論文名稱: 新生期投與dexamethasone對母鼠的長期不良影響
Long-term adverse effects of neonatal dexamethasone treatment in female rat
指導教授: 呂國棟
Lu, Kwok-Tung
楊奕玲
Yang, Yi-Ling
學位類別: 博士
Doctor
系所名稱: 生命科學系
Department of Life Science
論文出版年: 2018
畢業學年度: 106
語文別: 英文
論文頁數: 180
中文關鍵詞: 醣皮質激素地塞米松 (dexamethasone)海馬迴杏仁核雌激素受體類憂鬱行為長期增益現象表觀遺傳學組蛋白乙醯化
英文關鍵詞: Glucocorticoid, Dexamethasone, Hippocampus, Amygdala, Estrogen receptor, Depression-like behavior, Long-term potentiation, Epigenetic, Histone acetylation
DOI URL: http://doi.org/10.6345/DIS.NTNU.SLS.024.2018.D01
論文種類: 學術論文
相關次數: 點閱:217下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 地塞米松 (dexamethasone, DEX) 是一種人工合成的醣皮質激素(synthetic glucocorticoid),目前廣泛做為消炎藥劑(anti-inflammatory),並用於治療早產兒之呼吸窘迫症 (respiratory distress syndrome),但多年來研究發現新生期投與DEX (neonatal DEX treatment; NDT) 會導致腦部發育與認知功能的不良影響。海馬迴 (hippocampus) 是空間記憶 (spatial memory) 和連結性學習記憶(associative learning and memory) 的主要腦區,而杏仁核 (amygdala)則是調控情緒的重要腦區,兩者均屬於邊緣系統 (limbic system) 的成員,其功能異常與精神疾病如憂鬱症 (major depression) 及焦慮症 (anxiety disorder) 有重要關聯。
    前人研究顯示NDT會導致大鼠的類憂鬱行為 (depression-like behavior) 明顯增加,過去關於NDT的研究多著重於公鼠,較少著墨於母鼠,本研究利用新生期母鼠,模擬臨床上早產兒的投藥方式,以濃度遞減式 (tapering dosage) 於出生後第1~3天 (postnatal day-1 to 3) 連續3天依序投予DEX (0.5 mg/kg, 0.3 mg/kg 和 0.1 mg/kg),並利用細胞外電刺激紀錄 (in vitro extracellular recording)、西方墨點法 (western blot analysis)、抑制逃避學習實驗 (inhibitory avoidance task; IA) 與強迫游泳行為實驗 (forced swimming tests; FST),評估NDT對青春期 (6週齡) 與成年期 (10週齡) 的母鼠海馬迴與杏仁核之功能影響。
    實驗結果發現在海馬迴和杏仁核,利用高頻電刺激 (high-frequency stimulation; HFS) 誘發的長期增益現象 (long term potentiation; LTP) 中,青春期NDT母鼠的LTP有抑制的情形,而在成年期母鼠的海馬迴和杏仁核以HFS誘發的LTP則有減弱的情形。西方墨點法的結果顯示青春期與成年期母鼠海馬迴與杏仁核的estrogen receptor alpha (ER) 表現量均低於控制組,而estrogen receptor beta (ER) 的表現量與控制組無顯著差異。在檢測海馬迴空間記憶功能的IA行為實驗發現,NDT阻斷了青春期NDT母鼠抑制逃避學習,青春期NDT母鼠滯留潛伏 (retention latencies) 時間低於控制組並有顯著差異,但這種阻斷效果不會持續到成年期。評估杏仁核功能的FST行為實驗發現,青春期與成年期母鼠在FST模式中,不游動時間百分比 (percent time of immobility) 明顯高於控制組,表現出類憂鬱行為的反應增加。為避免NDT所引起的可能的毒性效果 (toxic effect) 對行為實驗數據判讀的干擾,故合併測量其自發性活動行為 (locomotor activity),結果顯示NDT母鼠在青春期和成年期的總水平移動距離 (total horizontal distance movement) 與控制組相比,並無顯著差異。在青春期和成年期母鼠在細胞外電生理紀錄時投予雌二醇 (estradiol) 可部分恢復杏仁核的HFS誘發之LTP;青春期動物於6週齡時連續四天皮下注射雌二醇 (0.25 mg/kg/day),成年期動物於10週齡時連續四天皮下注射雌二醇,分別在第四天給藥結束30分鐘後立即進行FST行為實驗,結果發現給予雌二醇的NDT母鼠,其類憂鬱行為降低。青春期母鼠在細胞外電生理紀錄給予雌二醇可部分恢復海馬迴的LTP。在染色質免疫沉澱實驗 (chromatin immunoprecipitation assay; ChIP) 中發現幼年期NDT母鼠 (2週齡) 的海馬迴中H3K9乙醯化 (acetylation) 與控制組相比,NDT母鼠的H3K9乙醯化百分比顯著低於控制組。在NDT組於出生後第一天 同時投與histone deacetylases (HDAC) 抑制劑trichostatin A (TSA),可使幼年期母鼠海馬迴ER的表現量恢復至與控制組相近,並可使青春期母鼠海馬迴以HFS誘發的LTP恢復,與控制組相比無顯著差異,以及在IA行為實驗發現海馬迴空間性記憶功能恢復,與控制組相比無顯著差異。總結本篇實驗結果,NDT母鼠在青春期與成年期的海馬迴與杏仁核之LTP、ER表現量、空間記憶及情緒功能均產生不良的影響,然而這樣不利的影響會一直持續到成年期,並且這樣的影響可能與H3K9乙醯化下降有關,而於幼年期投予HDAC抑制劑,或於青春期與成年期給予雌二醇,則可有效地降低NDT的長期不良影響。根據本論文的結果發現,NDT對母鼠產生的長期不良影響並直至成年期,這樣的結果可提供給日後相關研究及臨床投藥做為參考。

    Synthetic glucocorticoid dexamethasone (DEX) has been widely used as an anti-inflammatory agent and also used to treat respiratory distress syndrome in preterm infants. Previous studies suggested that neonatal DEX treatment (NDT) could alter brain development and cognitive function. Hippocampus is a critical brain area for both spatial and associative learning and memory. Amygdala is an essential region for regulating emotional response and behavior. Both hippocampus and amygdala are also the important components of the limbic system, which exhibit a tight association with mental disorders such as major depression and anxiety disorder. Previous NDT studies often focused on its influence with the male rats only. Because females are more vulnerable to the psychological stress and have higher susceptibility of having mental disorders, it is important and worthy to study the possible long-term adverse effect on female animals. In order to investigate the effects of NDT on the hippocampal and amygdaloid function in female rats, the experimental subjects received subcutaneous injections of a tapering dose of DEX (0.5 mg/kg, 0.3 mg/kg and 0.1 mg/kg) from postnatal day 1 to day 3 (PND1~3). In vitro electrophysiological recording, inhibitory avoidance (IA) and forced swimming test (FST) were used to evaluate the hippocampal and amygdaloid function, particularly the spatial and emotional memory function at juvenile (6 weeks old) and adult (10 weeks old) female rats. We found that NDT could alter high frequency stimulation (HFS) which would induce hippocampal and amygdaloid long-term potentiation (LTP) formation. The expression level of hippocampal and amygdaloid estrogen receptor alpha (ER) was significantly repressed in NDT female rats. The results of this study also showed that NDT blocked IA learning in juvenile NDT female rats which did not sustain to the adulthood. No significant difference in the retention latencies was found between NDT and SAL groups in adult female rats. The long-term adverse effects of NDT on amygdaloid function in both juvenile and adult NDT female rats were investigated by forced swimming test (FST). The FST results demonstrated that the depression-like behaviors were increased in both juvenile and adult NDT female rats as compared with the control group of SAL animals. By using locomotor activity monitoring, we can also study the possible toxic effect of NDT on motor function and spontaneous behavior. The total horizontal distance movement did not reveal any significant difference between the NDT and the control animals.
    Clearly, the normal HFS induced long-term potentiation (LTP) formation in hippocampus and amygdala would be blocked in juvenile and adult NDT animals. Suprafusion of estradiol could partially restore the hippocampal and amygdaloid LTP formation in NDT female rats, as the systemic administration (6 weeks old or 10 weeks old consecutive four days; 0.25 mg/kg/day; subcutaneous injection) of estradiol could partially attenuate depression-like behavior in both juvenile and adult NDT female rats. More interestingly, our chromation immunoprecipitation (ChIP) results revealed that the level of H3K9 acetylation was significantly decreased in the hippocampus of NDT female rats. Co-administration of HDAC inhibitor trichostatin A (TSA) could restore the expression of hippocampal ER expression in NDT female rats. In addition, TSA was also found to partially restore the hippocampal LTP formation and rescue the hippocampal spatial memory function on IA test in the NDT juvenile female rats.
    In conclusion, our results strongly suggested that NDT could elicit some long-term adverse effects on the hippocampus and amygdala in NDT female rats, and it is correlated to the decline of H3K9 acetylation, which resulted in the decrease of hippocampal and amygdaloid ER expression. Co-administration of TSA or systemic injection of estradiol could partially restore the hippocampal and amygdaloid function in NDT female rats. These results suggested that the hippocampal and amygdaloid level is closely related to the mechanism that causes ERα to incur the adverse effect of NDT female rats, which might result in adverse consequences in adult age.

    致謝 -1 Table of content -3 Abbreviation-10 Abstract in Chinese -13 Abstract in English -17 Introduction -20 1. HPA axis and the clinical usage of synthetic glucocorticoids -20 2. Neonatal dexamethasone treatment clinical application and its long-term adverse effect -23 3. The role of glucocorticoids in hypothalamic-pituitary-gonadal axis -27 4. Function of limbic system (hippocampus & amygdala) -30 a. The functional role of hippocampus -31 b. The functional role of amygdala in emotion -33 5. The functional of 17 β-estradiol and estrogen receptor -34 6. Epigenetic, its biological role, and epigenetic modulation of the estrogen receptors -38 Materials and methods -43 Subjects -43 Neonatal dexamethasone administration protocol -43 Trichostatin A administration -44 In vitro and in vivo estradiol administration -45 a. For extracellular recording -45 b. For behavioral experiment -45 Brain slice extracellular recording -45 Inhibitory avoidance task -47 Forced swimming tests -48 Locomotor activity test -49 Western blot analysis -49 Chromatin immunoprecipitation -51 Vaginal secretion test -54 Enzyme linked immunosorbent assays -56 Statistical analysis -57 Results of chapter 1 -58 Experiment 1-1 -59 Aim -59 Rationale -59 Procedure -59 Result-60 Experiment 1-2 -60 Aim -60 Rationale -60 Procedure -61 Result -61 Experiment 1-3 -62 Aim -62 Rationale -62 Procedure -62 Result -63 Experiment 1-4 -64 Aim -64 Rationale -64 Procedure -64 Result -64 Experiment 1-5 -65 Aim -65 Rationale -65 Procedure -66 Result -66 Experiment 1-6 -67 Aim -67 Rationale -67 Procedure -68 Result -68 Results of chapter 2 -70 Experiment 2-1 -71 Aim -71 Rationale -71 Procedure -71 Result -72 Experiment 2-2 -73 Aim -73 Rationale -73 Procedure -74 Result -74 Experiment 2-3 -75 Aim -75 Rationale -75 Procedure -76 Result -76 Experiment 2-4 -77 Aim -77 Rationale -77 Procedure -78 Result -78 Experiment 2-5 -79 Aim -79 Rationale -79 Procedure -79 Result -80 Experiment 2-6 -81 Aim -81 Rationale -81 Procedure -81 Result -82 Experiment 2-7 -82 Aim -82 Rationale -82 Procedure -83 Result -83 Results of chapter 3 -85 Experiment 3-1 -86 Aim -86 Rationale -86 Procedure -86 Result -86 Experiment 3-2 -87 Aim -87 Rationale -87 Procedure -87 Result -88 Experiment 3-3 -89 Aim -89 Rationale -89 Procedure -90 Result -90 Experiment 3-4 -91 Aim -91 Rationale -91 Procedure -92 Result -92 Experiment 3-5 -93 Aim -93 Rationale -93 Procedure -93 Result -94 Experiment 3-6 -95 Aim -95 Rationale -95 Procedure -96 Result -96 Discussion and conclusion -99 References -114 Figures -143

    Adams, R.D. and Victor, M., Principles of Neurology, 3rd ed., MacGraw-Hill, New York, 1985.
    Azcoitia I, Sierra A, Garcia-Segura LM. Localization of estrogen receptor beta-immunoreactivity in astrocytes of the adult rat brain. Glia. 1999, 26, 260-267.
    Aisa B, Tordera R, Lasheras B, Del Río J, Ramírez MJ. Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrino. 2007, 32, 256-266.
    Aisa B, Gil-Bea FJ, Marcos B, Tordera R, Lasheras B, Del Río J, et al., Neonatal stress affects vulnerability of cholinergic neurons and cognition in the rat: involvement of the HPA axis. Psychoneuroendocrino. 2009, 34, 1495-1505.
    Akhoundi-Khalafi AM, Shishehbore MR. A new technique for quantitative determination of dexamethasone in pharmaceutical and biological samples using kinetic spectrophotometric method. Int J Anal Chem. 2015, 439271.
    Álvarez-Errico D, Vento-Tormo R, Sieweke M, Ballestar E. Epigenetic control of myeloid cell differentiation, identity and function. Nat Rev Immunol. 2015, 15, 7-17.
    Abaya R, Jones L, Zorc JJ. Dexamethasone Compared to Prednisone for the Treatment of Children With Acute Asthma Exacerbations. Pediatr Emerg Care. 2018, 34, 53-58.
    Burri PH. The postnatal growth of the rat lung. III. Morphology. Anat Rec. 1974, 80, 77-98.
    Briski KP, Sylvester PW. Acute inhibition of pituitary LH release in the male rat by the glucocorticoid agonist decadron phosphate. Neuroendocrinology. 1991, 54, 313-320.
    Burgess LH, Handa RJ. Chronic estrogen-induced alterations in adrenocorticotropin and corticosterone secretion, and glucocorticoid receptormediated functions in female rats. Endocrinology. 1992, 131, 1261-1269.
    Brown RW, Diaz R, Robson AC, Kotelevtsev YV, Mullins JJ, Kaufman MH, Seckl JR. he ontogeny of 11β-hydroxysteroid dehydrogenase type 2 and mineralicorticoid receptor gene expression reveal intricate control of glucocorticoid action in development. Endocrinology. 1996, 137, 794-797.
    Bakker JM, Kavelaars A, Kamphuis PJ, Cobelens PM, van Vugt HH, van Bel F, Heijnen CJ. eonatal dexamethasone treatment increases susceptibility to experimental autoimmune disease in adult rats. J Immunol. 2000, 165, 5932-5937.
    Bi R, Foy MR, Vouimba RM, Thompson RF, Baudry M. Cyclic changes in estradiol regulate synaptic plasticity through the MAP kinase pathway. PNAS. 2001, 98, 13391-13395.
    Barrington KJ. Postnatal steroids and neurodevelopmental outcomes: a problem in the making. Pediatrics. 2001, 107, 1425-1426.
    Barrington KJ. The adverse neuro-developmental effects of postnatal steroids in the preterm infant: a systematic review of RCTs. BMC Pediatr. 2001, 1, 1
    Bantignies F, Grimaud C, Lavrov S, Gabut M, Cavalli G. Inheritance of polycomb dependent chromosomal interactions in drosophila. Genes & Development. 2003, 17, 2406-2420.
    Bourbon JR, Boucherat O, Boczkowski J, Crestani B, Delacourt C. Bronchopulmonary dysplasia and emphysema: in search of common therapeutic targets. Trends Mol Med. 2009,15, 169-179.
    Bertrand P. Inside HDAC with HDAC inhibitors. Eur J Med Chem. 2010, 45, 2095-2116.
    Barneda-Zahonero B, Parra M. Histone deacetylases and cancer. Mol Oncol. 2012, 6, 579-589.
    Becker DE. Basic and clinical pharmacology of glucocorticosteroids. Anesth Prog. 2013, 60, 25-32.
    Bellavance MA, Rivest S. The HPA-immune axis and the the immunomodulatory actions of glucocorticoids in the brain. Front Immunol. 2014, 31,136.
    Bean LA, Ianov L, Foster TC. Estrogen receptors, the hippocampus, and memory. Neuroscientist. 2014, 20, 534-545.
    Bowers ME, Ressler KJ. Sex-dependence of anxiety-like behavior in cannabinoid receptor 1 (Cnr1) knockout mice. Behav Brain Res. 2016, 300, 65-69.
    Baranowski ES, Arlt W, Idkowiak J. Monogenic Disorders of Adrenal Steroidogenesis. Horm Res Paediatr. 2018, 89, 292-310.
    Collu R, Gibb W, Ducharme JR. Effects of stress on the gonadal function. J Endocrinol Invest. 1984, 7, 529-537.
    Córdoba Montoya DA, Carrer HF. Estrogen facilitates induction of long term potentiation in the hippocampus of awake rats. Brain Res. 1997, 778, 430-438.
    Cardoso WV, Williams MC. Basic mechanisms of lung development: Eighth Woods Hole Conference on lung cell biology 2000. Am J Respir Cell Mol Biol. 2001, 25, 137-140.
    Cardoso WV. Molecular regulation of lung development. Annu Rev Physiol. 2001, 63, 471-494.
    Canonaco M, Facciolo RM, Alo R. Neuroactive steroid mechanisms and GABA type A receptor subunit assembly in hypothalamic and extrahypothalamic regions. Int Rev Cytol. 2002, 214, 63-101.
    Cohen AW, Hnasko R, Schubert W, Lisanti MP. Role of caveolae and caveolins in health and disease. Physiol Rev. 2004, 84, 1341-1379.
    Cooke BM, Woolley CS. Gonadal hormone modulation of dendrites in the mammalian CNS. J Neurobiol. 2005, 64, 34-46.
    Canonaco M, Giusi G, Madeo A, Facciolo RM, Lappano R, Canonaco A, et al., A sexually dimorphic distribution pattern of the novel estrogen receptor G-protein-coupled receptor 30 in some brain areas of the hamster. J Endocrinol. 2008, 196, 131-138.
    Cui J, Shen Y, Li R. Estrogen synthesis and singaling pathway during aging: form periphery to brain. Trends Mol Med. 2013, 19, 197-209.
    Cao Y, Wu R, Tai F, Zhang X, Yu P, An, X, Qiao X, Hao P. Neonatal paternal deprivation impairs social recognition and alters levels of oxytocin and estrogen receptor α mRNA expression in the MeA and NAcc, and serum oxytocin in mandarin voles. Horm Behav. 2014, 65, 57-65.
    Cheong JLY, Burnett AC, Lee KJ, Roberts G, Thompson DK, Wood SJ, Connelly A, Anderson PJ, Doyle LW, Victorian Infant Collaborative Study Group. Association between postnatal dexamethasone for treatment of bronchopulmonary dysplasia and brain Volumes at Adolescence in Infants Born Very Preterm. J Pediatr. 2014, 164, 737-743.
    Cain DW, Cidlowski JA. Immune regulation by glucocorticoids. Nat Rev Immunol. 2017, 17, 233-247.
    Dubey AK, Plant TM. A suppression of gonadotropin secretion by cortisol in castrated male rhesus monkeys (Macaca mulatta) mediated by the interruption of hypothalamic gonadotropin-releasing hormone release. Biol Reprod. 1985, 33, 423-431.
    Davis EP, Bruce J, Gunnar MR. The anterior attention network: associations with temperament and neuroendocrine activity in 6-year-old children. Dev Psychobiol. 2002, 40, 43-56.
    Drapeau E, Mayo W, Aurousseau C, Le Moal M, Piazza PV, Abrous DN. Spatial memory performances of aged rats in the water maze predict levels of hippocampal neurogenesis. Proc Natl Acad Sci USA. 2003, 100, 14385-14390.
    Dokmanovic M, Marks PA. Prospects: histone deacetylase inhibitors. J Cell Biochem. 2005, 96, 293-304.
    Dokmanovic M, Clarke C, Marks PA. Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res. 2007, 5, 981-989.
    Davis EP, Sandman CA. The timing of prenatal exposure to maternal cortisol and psychosocial stress is associated with human infant cognitive development. Child Dev. 2010, 81, 131-148.
    de Vries WB, van den Borne P, Goldschmeding R, de Weger RA, Bal MP, van Bel F, et al., Neonatal dexamethasone treatment in the rat leads to kidney damage in adulthood. Pediatr Res. 2010, 67, 72-76.
    Demauro SB, Dysart K, Kirpalani H. Stopping the swinging pendulum of postnatal corticosteroid use. J Pediatr. 2014, 164, 9-11.
    D'Urso A, Brickner JH. Mechanisms of epigenetic memory. Trends Genet. 2014, 30, 230-236.
    Duarte-Guterman P, Yagi S, Chow C, Galea L A. Hippocampal learning, memory, and neurogenesis: effects of sex and estrogens across the lifespan in adults. Horm Behav. 2015, 74, 37-52.
    Entringer S, Kumsta R, Hellhammer DH, Wadhwa PD, Wüst S. Prenatal exposure to maternal psychosocial stress and HPA axis regulation in young adults. Horm Behav. 2009, 55, 292-298.
    Fauser A, Pohlandt F, Bartmann P, Gortner L. Rapid increase of blood pressure in extremely low birth weight infants after a single dose of dexamethasone. Eur J Pediatr. 1993, 152, 354-356.
    Flagel SB, Vázquez DM, Watson SJ Jr., Neal CR Jr. Effects of tapering neonatal dexamethasone on rat growth, neurodevelopment, and stress response. Am J Physiol-Reg I. 2002, 282, R55-R63.
    Frye CA, Walf AA. Estrogen and/or progesterone administered systemically or to the amygdala can have anxiety-, fear-, and pain-reducing effects in ovariectomized rats. Behav Neurosci. 2004, 118, 306-313.
    Filardo EJ, Thomas P, GPR30: a seven-transmembrane-spanning estrogen receptor that triggers EGF release. Trends Endocrinol Metab. 2005, 16, 362-367.
    Foy MR, Baudry M, Diaz Brinton R, Thompson RF. Estrogen and hippocampal plasticity in rodent models. J Alzheimers Dis. 2008, 15, 589-603.
    Foy MR, Ovarian hormones, aging and stress on hippocampal synaptic plasticity. Neurobiol Learn Mem. 2011, 95, 134-144.
    Furuta M, Numakawa T, Chiba S, Ninomiya M, Kajiyama Y, Adachi N, et al., Estrogen, predominantly via estrogen receptor α, attenuates postpartum-induced anxiety- and depression-like behaviors in female rats. Endocrinology. 2013, 154, 3807-3816.
    Fester L, Rune GM. Sexual neurosteroids and synaptic plasticity in the hippocampus. Brain Res. 2015, 1621, 162-169.
    Gilstrap LC, Christensen R, Clewell WH, D'Alton ME, Davidson Jr EC, Escobedo MB, Gjerdingen DK, Goddard-Finegold J, Goldenberg RL, Grimes DA, Hansen TN, Kauffman RE, Keeler EB, Oh W, Susman EJ, Vogel MG, Avery ME, Ballard PL, Ballard RA, Crowley P, Garite T, Hankins GDV, Jobe AH, Koppe JG, Maher JE, Merkatz IR, Shankaran S, Simpson KN, Sinclair JC, Slotkin TA, Taeusch Jr HW, Wright LL, Alexander D, Berberich MA, Bracken M, Cooper L, Culpepper L, Elliott JM, Ferguson JH, Frigoletto F, Gail DB, Hall WH, M. Jones Jr D, Medoff-Cooper B, Merenstein GB, Whalen JM, Lenfant C, Hinshaw AS. Effect of corticosteroids for fetal maturation on perinatal outcomes. NIH consensus development panel on the effect of corticosteroids for fetal maturation on perinatal outcomes. JAMA. 1995, 273, 413-418.
    Gill AW, Warner G, Bull L. Iatrogenic neonatal hypertrophic cardiomyopathy. Pediatr Cardiol. 1996, 17, 335-339.
    Gordon PV, Young ML, Marshall DD. Focal small bowel perforation: an adverse effect of early postnatal dexamethasone therapy in extremely low birth weight infants. J Perinatol. 2001, 21, 156-160.
    Gavranidou M, Rosner R. The weaker sex? Gender and post-traumatic stress disorder. Depress Anxiety. 2003, 17, 130-139.
    Goel N, Bale T. Organizational and activational effects of testosterone on masculinization of female physiological and behavioral stress responses. Endocrinology. 2008, 149, 6399-6405.
    Goel N, Bale TL. Sex differences in the serotonergic influence on the hypothalamic-pituitary-adrenal stress axis. Endocrinology. 2010, 151, 1784-1794.
    Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, et al., Histone methylation regulates memory formation. J Neurosci. 2010, 30, 3589-3599.
    Gupta-Agarwal S, Franklin AV, Deramus T, Wheelock M, Davis RL, McMahon LL, Lubin FD. G9a/GLP histone lysine dimethyltransferase complex activity in the hippocampus and the entorhinal cortex is required for gene activation and silencing during memory consolidation. J Neurosci. 2012, 32, 5440-5453.
    Galea LA, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol. 2013, 25, 1039-1061.
    Goel N, Workman JL, Lee TT, Innala L, Viau V. Sex differences in the HPA axis. Compr Physiol. 2014, 4, 1121-1155.
    Geraghty AC, Muroy SE, Zhao S, Bentley GE, Kriegsfeld LJ, Kaufer D. Knockdown of hypothalamic RFRP3 prevents chronic stress-induced infertility and embryo resorption. Elife. 2015, 12, 4.
    Geraghty AC, Kaufer D. Glucocorticoid Regulation of Reproduction. Adv Exp Med Biol. 2015, 872, 253-278.
    Handa RJ, Burgess LH, Kerr JE, O’Keefe JA. Gonadal steroid hormone receptors and sex differences in the hypothalamo-pituitary-adrenal axis. Horm Behav. 1994, 28, 464-476.
    Hosokawa K, Ottander U, Wahlberg P, Ny T, Cajander S, Olofsson IJ. Dominant expression and distribution of oestrogen receptor beta over oestrogen receptor alpha in the human corpus luteum. Mol Hum Reprod. 2001, 7, 137-145.
    He J, Crews FT. Neurogenesis decreases during brain maturation from adolescence to adulthood. Pharmacol Biochem Behav. 2007, 86, 327-333.
    Hodes GE, Yang L, VanKooy J, Santollo J, Shors TJ. Prozac during puberty: distinctive effects on neurogenesis as a function of age and sex. Neuroscience. 2009, 163, 609-617.
    Hagelkruys A, Sawicka A, Rennmayr M, Seiser C. The biology of HDAC in cancer: the nuclear and epigenetic components. Handb Exp Pharmacol. 2011, 206, 13-37.
    Herriges M, Morrisey EE. Lung development: orchestrating the generation and regeneration of a complex organ. Development. 2014, 141, 502-513.
    Handa RJ, Weiser MJ. Gonadal steroid hormones and the hypothalamo-pituitary-adrenal axis. Front Neuroendocrinol. 2014, 35, 197-220.
    lversen, S.D., Recent advances in the anatomy and chemistry of the limbic system. In M.R. Trimble and E. Zarifian (Eds.), Psychophannacology of the Lhnbic System, Oxford University Press, Oxford. 1984, 1-16.
    Insel TR. Regional changes in brain oxytocin receptors post-partum: time-course and relationship to maternal behaviour. J Neuroendocrinol. 1990a, 2, 539-545.
    Insel TR. Regional induction of c-fos-like protein in rat brain after estradiol administration. Endocrinology. 1990b, 126, 1849-1853.
    Jenuwein T, Allis CD. Translating the histone code. Science. 2001, 293, 1074-1080.
    Johnson PH, Walker RP, Jones SW, Stephens K, Meurer J, Zajchowski DA, et al., Multiplex gene expression analysis for high-throughput drug discovery: screening and analysis of compounds affecting genes overexpressed in cancer cells. Mol Cancer Ther. 2002, 1, 1293-1304.
    Jelks KB, Wylie R, Floyd CL, McAllister AK, Wise P, Estradiol targets synaptic proteins to induce glutamatergic synapse formation in cultured hippocampal neurons: critical role of estrogen receptor-alpha. J Neurosci. 2007, 27, 6903-6913.
    Johnson RL, Huang W, Jadhav A, Austin CP, Inglese J, Martinez ED. A quantitative high-throughput screen identifies potential epigenetic modulators of gene expression. Anal Biochem. 2008, 375, 237-248.
    Jablonka E, Raz G. Transgenerational epigenetic inheritance: prevalence, mechanisms, and implications for the study of heredity and evolution. Q Rev Biol. 2009, 84, 131-176.
    Jeong EA, Jeon BT, Kim JB, Kim JS, Cho YW, Lee DH, Kim HJ, Kang SS, Cho GJ, Choi WS, Roh GS. Phosphorylation of 14-3-3ζ at serine 58 and neurodegeneration following kainic acid induced excitotoxicity. Anat Cell Biol. 2010, 43, 150-156.
    Jefferies JM, Cooper T, Yam T, Clarke SC. Pseudomonas aeruginosa outbreaks in the neonatal intensive care unit - a systematic review of risk factors and environmental sources. J Med Microbiol. 2012, 61, 1052-1061.
    Jarome TJ, Lubin FD. Histone lysine methylation: Critical regulator of memory and behavior. Rev. Neurosci. 2013, 24, 375-387.
    Jie Cui, Yong Shen, Rena Li. Estrogen synthesis and signaling pathways during ageing: from periphery to brain. Trends Mol Med. 2013, 19, 197-209.
    Kamel F, Kubajak CL. Modulation of gonadotropin secretion by corticosterone: interaction with gonadal steroids and mechanism of action. Endocrinology. 1987, 121, 561-568.
    Kari MA, Hallman M, Eronen M, Teramo K, Virtanen M, Koivisto M. Ikonen RS, Prenatal dexamethasone treatment in conjunction with rescue therapy of human surfactant: a randomized placebo-controlled multicenter study. Pediatrics. 1994, 93, 730-736.
    Kirschbaum C, Schommer N, Federenko I, Gaab J, Neumann O, Oellers M, Rohleder N, Untiedt A, Hanker J, Pirke KM, Hellhammer DH. Short-term estradiol treatment enhances pituitary-adrenal axis and sympathetic responses to psychosocial stress in healthy young men. J Clin Endocrinol Metab. 1996, 81, 3639-3643.
    Klar AJ. Propagating epigenetic states through meiosis: where Mendel's gene is more than a DNA moiety. Trends Genet. 1998, 14, 299-301.
    Kandel ER, Schwartz JH, Jessell TM. Principles of Neural Science. McGraw-Hill, 2000.
    Kamphuis PJ, Gardoni F, Kamal A, Croiset G, Bakker JM, Cattabeni F, et al., Long-lasting effects of neonatal dexamethasone treatment on spatial learning and hippocampal synaptic plasticity: involvement of the NMDA receptor complex. FASEB J. 2003, 17, 911-913.
    Kouzarides T. Chromatin modifications and their function. Cell. 2007, 128, 693-705.
    Kiefer J. Epigenetics in development - Kiefer - 2007 - Developmental Dynamics - Wiley Online Library. Developmental dynamics, 2007.
    Kim HJ, Bae SC. Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res. 2011, 3, 166-179.
    Karmakar S, Jin Y, Nagaich AK. Interaction of glucocorticoid receptor (GR) with estrogen receptor (ER) α and activator protein 1 (AP1) in dexamethasone-mediated interference of ER activity. J Biol Chem. 2013, 288, 24020-24034.
    Ko MC, Hung YH, Ho PY, Yang YL, Lu KT. Neonatal glucocorticoid treatment increased depression-like behaviour in adult rats. Int J Neuropsychoph. 2014a, 17, 1995-2004.
    Ko MC, Lee MC, Amstislavskaya TG, Tikhonova MA, Yang YL, Lu KT. Inhibition of NKCC1 attenuated hippocampal LTP formation and inhibitory avoidance in Rat. PLoS One. 2014b, 9, e106692.
    le Cras TD, Markham NE, Morris KG, Ahrens CR, McMurtry IF. Abman SH, Neonatal dexamethasone treatment increases the risk for pulmonary hypertension in adult rats. Am J Physiol-Lung C. 2000, 278, L822-L829.
    Lemaire V, Koehl M, LeMoal M, Abrous DN. Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus. Proc Natl Acad Sci USA. 2000, 97, 11032-11037.
    LeFlore JL, Salhab WA, Broyles RS, Engle WD. Association of antenatal and postnatal dexamethasone exposure with outcomes in extremely low birth weight neonates. Pediatrics. 2002, 110, 275-279.
    Lowry CA. Functional subsets of serotonergic neurones: implications for control of the hypothalamic-pituitary-adrenal axis. J Neuroendocrinol. 2002, 14, 911-923.
    Luine VN, Jacome LF, Maclusky NJ. Rapid enhancement of visual and place memory by estrogens in rats. Endocrinology. 2003, 144, 2836-2844.
    Lamprecht R, LeDoux J. Structural plasticity and memory. Nat Rev Neurosci. 2004, 5, 45-54.
    Lund TD, Rovis T, Chung WC, Handa RJ. Novel actions of estrogen receptor-beta on anxiety-related behaviors. Endocrinology. 2005, 146, 797-807.
    Lee TI, Johnstone SE, Young RA. Chromatin immunoprecipitation and microarray-based analysis of protein location. Nat Protoc. 2006, 1, 729-748.
    Lin HJ, Huang CC, Hsu KS. Effects of neonatal dexamethasone treatment on hippocampal synaptic function. Ann Neurol. 2006, 59, 939-951.
    Lee BH, Stoll BJ, McDonald SA, Higgins RD. Neurodevelopmental outcomes of extremely low birth weight infants exposed prenatally to dexamethasone versus betamethasone. Pediatrics. 2008, 121, 289-296.
    Lupien SJ, McEwen BS, Gunnar MR, Heim C. Effects of stress throughout the life span on the brain, behaviour and cognition. Nat Rev Neurosci. 2009, 10, 434-445.
    Lubin FD. Epigenetic gene regulation in the adult mammalian brain: multiple roles in memory formation. Neurobiol Learn Mem. 2011, 96, 68-78.
    Lubin FD, Gupta S, Parrish RR, Grissom NM, Davis RL. Epigenetic mechanisms: critical contributors to long-term memory formation. Neuroscientist. 2011, 17, 616-632.
    Li Y, Meeran SM, Patel SN, Chen H, Hardy TM, Tollefsbol TO. Epigenetic reaction of estrogen receptor-α (ERα) by genistein enhances hormonal therapy sensitivity in ERα-negative breast cancer. Mol Cancer. 2013, 12.
    Lucena GM, Prediger RD, Silva MV, Santos SN, Silva JF, Santos AR, Azevedo MS, Ferreira VM. Ethanolic extract from bulbs of Cipura paludosa reduced long-lasting learning and memory deficits induced by prenatal methylmercury exposure in rats. Dev Cogn Neurosci. 2013, 3, 1-10.
    Liu SB, Tian Z, Guo YY, Zhang N, Feng B, Zhao MG. Activation of GPR30 attenuates chronic pain-related anxiety in ovariectomized mice. Psychoneuroendocrinology. 2015, 53, 94-107.
    Lu J, Wu XY, Zhu QB, Li J, Shi LG, Wu JL, Zhang QJ, Huang ML, Bao AM. Sex differences in the stress response in SD rats. Behav Brain Res. 2015, 284, 231-237.
    Lanshakov DA, Sukhareva EV, Kalinina TS, Dygalo NN. Dexamethasone-induced acute excitotoxic cell death in the developing brain. Neurobiol Dis. 2016, 91, 1-9.
    Leibel S, Post M. Endogenous and exogenous stem/progenitor cells in the lung and their role in the pathogenesis and treatment of pediatric lung disease. Front Pediatr. 2016, 14, 36.
    Massaro D, Teich N, Maxwell S, Massaro GD, Whitney P. Postnatal development of alveoli: Regulation and evidence for a critical period in rats. J Clin Invest. 1985, 76, 1297-1305.
    Meyer ME, Gronemeyer H, Turcotte B, Bocquel MT, Tasset D, Chambon P. Steroid hormone receptors compete for factors that mediate their enhance function. Cell. 1989, 57, 433-442.
    Menasce LP, White GR, Harrison CJ, Boyle JM. Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-FISH banding technique. Genomics. 1993, 17, 263-265.
    Murphy DD, Cole NB, Greenberger V, Segal M. Estradiol increases dendritic spine density by reducing GABA neurotransmission in hippocampal neurons. J Neurosci. 1998, 18, 2550-2559.
    McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999, 20, 279-307.
    Micheva-Viteva S, Kobayashi Y, Edelstein LC, Pacchia AL, Lee HL, Graci JD, et al., High-throughput Screening Uncovers a Compound That Activates Latent HIV-1 and Acts Cooperatively with a Histone Deacetylase (HDAC) Inhibitor. J Biol Chem. 2001, 286, 21083-21091.
    Marcondes FK, Bianchi FJ, Tanno AP. Determination of the estrous cycle phases of rat: some helpful considerations. Braz J Bio. 2002, 62, 609-614.
    McEwen BS. Early life influences on life-long patterns of behavior and health. Ment Retard Dev D R. 2003, 9, 149-154.
    Miller TA, Witter DJ, Belvedere S. Histone deacetylase inhibitors. J Med Chem. 2003, 46, 5097-5116.
    Mitra SW, Hoskin E, Yudkovitz J, Pear L, Wilkinson HA, Hayashi S, et al., Immunolocalization of estrogen receptor beta in the mouse brain: comparison with estrogen receptor alpha. Endocrinology. 2003, 144, 2055-2067.
    Murphy VE, Clifton VL. Alterations in human placental 11beta-hydroxysteroid dehydrogenase type 1 and 2 with gestational age and labour. Placenta. 2003, 24, 739-744.
    Mitsushima D, Masuda J, Kimura F. Sex differences in the stressinduced release of acetylcholine in the hippocampus and corticosterone from the adrenal cortex in rats. Neuroendocrinology. 2003, 78, 234-240.
    Macaluso M, Cinti C, Russo G, Russo A, Giordano A. pRb2 /p130-E2F4 /5-HDAC1-SUV39H1-p300 and pRb2 /p130-E2F4 /5-HDAC1 -SUV39H1-DNMT1 multimolecular complexes mediate the transcription of estrogen receptor-alpha in breast cancer. Oncogene. 2003, 22, 3511-3517.
    Moore PS, Barbi S, Donadelli M, Costanzo C, Bassi C, Palmieri M, Scarpa A. Gene expression profiling after treatment with the histone deacetylase inhibitor trichostatin A reveals altered expression of both pro- and anti-apoptotic genes in pancreatic adenocarcinoma cells. Biochim Biophys Acta. 2004, 1693, 167-176.
    Montaron MF, Drapeau E, Dupret D, Kitchener P, Aurousseau C, Le Moal M, et al. Lifelong corticosterone level determines age-related decline in neurogenesis and memory. Neurobiol Aging. 2006, 27, 645-654.
    Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. 2006, 6, 38-51.
    Marais L, van Rensburg SJ, van Zyl JM, Stein DJ, Daniels WM. Maternal separation of rat pups increases the risk of developing depressive-like behavior after subsequent chronic stress by altering corticosterone and neurotrophin levels in the hippocampus. Neurosci Res. 2008, 61, 106-112.
    Mill J, Kiss E, Baji I, Kapornai K, Daróczy G, Vetró A, et al., Association study of the Estrogen Receptor Alpha Gene (ESR1) and childhood-onset mood disorders. Am J Med Genet B. 2008, 147B, 1323-1326.
    McGowan PO, Sasaki A, D'Alessio AC, Dymov S, Labonté B, Szyf M, Turecki G, Meaney MJ. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci. 2009, 12, 342-348.
    Maeng LY, Milad MR. Sex differences in anxiety disorders: Interactions between fear, stress, and gonadal hormones. Horm Behav. 2015, 76, 106-117.
    McEwen BS, Nasca C, Gray JD. Stress Effects on Neuronal Structure: Hippocampus, Amygdala, and Prefrontal Cortex. Neuropsychopharmacol. 2016, 41, 3-23.
    Nishizuka M, Arai Y. Synapse formation in response to estrogen in the medial amygdala developing in the eye. Proc Natl Acad Sci U S A. 1982, 79, 7024-7026.
    Nieuwenhuys R, Voogd J, van Huijzen C. The Human Central Nervous System, Springer, Berlin, Germany, 1996.
    Nelson RJ. An introduction to behavioral endocrinology. Sinauer Associates. Inc. Publishers, 2000.
    Nakayama J, Rice JC, Strahl BD, Allis CD, Grewal SI. Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly. Science. 2001, 292, 110-113.
    Nolte J. The Human Brain: An Introduction to Its Functional Anatomy, 5th edition, Mosby, 2002.
    Neal CR Jr., Weidemann, G, Kabbaj M, Vázquez DM. Effect of neonatal dexamethasone exposure on growth and neurological development in the adult rat. Am J Physiol-Reg I. 2004, 287, R375-R385.
    Nebieridze N, Zhang XL, Chachua T, Velíšek L, Stanton PK, Velíšková J. β-Estradiol unmasks metabotropic receptor-mediated metaplasticity of NMDA receptor transmission in the female rat dentate gyrus. Psychoneuroendocrino. 2012, 37, 1845-1854.
    Oosterlaken-Dijksterhuis MA, Haagsman HP, van Golde LM, Demel RA. Characterization of lipid insertion into monomolecular layers mediated by lung surfactant proteins SP-B and SP-C. Biochemistry. 1991, 30, 10965-10971.
    Omar SA, DeCristofaro JD, Agarwal BI, LaGamma EF. Effect of prenatal steroids on potassium balance in extremely low birth weight neonates. Pediatrics. 2000, 106, 561-567.
    Osterlund MK, Witt MR, Gustafsson JA. Estrogen action in mood and neurodegenerative disorders: estrogenic compounds with selective properties-the next generation of therapeutics. Endocrine. 2005, 28, 235-242.
    Osterlund MK. Underlying mechanisms mediating the antidepressant effects of estrogens. Biochim Biophys Acta. 2010, 1800, 1136-1144.
    Pfaff D, Keiner M. Atlas of estradiol-concentrating cells in the central nervous system of the female rat. J Comp Neurol. 1973, 151, 121-158.
    Porsolt RD, Anton G, Blavet N, Jalfre M, Behavioural despair in rats: a new model sensitive to antidepressant treatments. Eur J Pharmacol. 1978, 47, 379-391.
    Packard MG, Teather LA. Intra-hippocampal estradiol infusion enhances memory in ovariectomized rats. Neuroreport. 1997a, 8, 3009-3013.
    Packard MG, Teather LA. Posttraining estradiol injections enhance memory in ovariectomized rats: cholinergic blockade and synergism. Neurobiol Learn Mem. 1997b, 68, 172-188.
    Pitkänen A, Stefanacci L, Farb CR, Go GG, LeDoux JE, Amaral DG. Intrinsic connections of the rat amygdaloid complex: projections originating in the lateral nucleus. J Comp Neurol. 1995, 356, 288-310.
    Purves D, Augustine GJ, Chikaraishi DM, Ehlers MD, Einstein G, Fitzpatrick D, William CH, Jarvis E, Katz LC, Kauer J, LaMantia AS, McNamara JO, Mooney RD, Nicolelis MAL, Purves D, Reinhart PH, Simon SA, Skene P, Voyvodic J, White LE, Williams M. Neuroscience, 3rd Ed. Sinauer, 2004.
    Prediger RD, Fernandes MS, Rial D, Wopereis S, Pereira VS, Bosse TS, Da Silva CB, Carradore RS, Machado MS, Cechinel-Filho V, Costa-Campos L. Effects of acute administration of the hydroalcoholic extract of mate tea leaves (Ilex paraguariensis) in animal models of learning and memory. J Ethnopharmacol. 2008, 120, 465-473.
    Pawluski JL, Brummelte S, Barha CK, Crozier TM, Galea LA. Effects of steroid hormones on neurogenesis in the hippocampus of the adult female rodent during the estrous cycle, pregnancy, lactation and aging. Front Neuroendocrinol. 2009, 30, 343-357.
    Phipps SM, Love WK, White T, Andrews LG, Tollefsbol TO. Retinoid-induced histone deacetylation inhibits telomerase activity in estrogen receptor-negative breast cancer cells. Anticancer Res. 2009, 29, 4959-4964.
    Pike CJ, Carroll JC, Rosario ER, Barron AM. Protective actions of sex steroid hormones in Alzheimer's disease. Front Neuroendocrin. 2009, 30, 239-258.
    Popoli M, Yan Z, McEwen BS, Sanacora G. The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci. 2011, 13, 22-37.
    Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERα) and beta (ERβ): subtype-selective ligands and clinical potential. Steroids. 2014, 90, 13-29.
    Phumsatitpong C, Moenter SM. Estradiol-Dependent Stimulation and Suppression of Gonadotropin-Releasing Hormone Neuron Firing Activity by Corticotropin-Releasing Hormone in Female Mice. Endocrinology. 2018, 159, 414-425.
    Ryan KJ. Biochemistry of aromatase: significance to female reproductive physiology. Cancer Res. 1982, 42, 3342s-3344s.
    Rabin D, Gold PW, Margioris AN, Chrousos GP. Stress and reproduction: physiologic and pathophysiologic interactions between the stress and reproductive axes. Adv Exp Med Biol. 1988, 245, 377-387.
    Riggs AD, Porter TN. Overview of epigenetic mechanisms. Cold Spring Harbor Monograph Archive. 1996, 32, 29-45.
    Rudick CN, Woolley CS. Estrogen regulates functional inhibition of hippocampal CA1 pyramidal cells in the adult female rat. J Neurosci. 2001, 21, 6532-6543.
    Rice JC, Allis CD. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curr Opin Cell Biol. 2001, 13, 263-273.
    Rasia-Filho AA, Fabian C, Rigoti KM, Achaval M. Influence of sex, estrous cycle and motherhood on dendritic spine density in the rat medial amygdala revealed by the Golgi method. Neuroscience. 2004, 126, 839-847.
    Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. N Engl J Med. 2005, 353, 1711-1723.
    Rand S. Swenson, M.D. Review of clinical and functional neuroscience-swenson. Dartmouth medical school, 2006.
    Roxo MR, Franceschini PR, Zubaran C, Kleber FD, Sander JW. The Limbic System Conception and Its Historical Evolution. Scientific World Journal. 2011, 11, 2428-2441.
    Ryan J, Scali J, Carrière I, Peres K, Rouaud O, Scarabin PY, Ritchie K, Ancelin ML. Estrogen receptor alpha gene variants and major depressive episodes. J Affect Disord. 2012, 136, 1222-1226.
    Rocca WA, Ulrich LG. Oophorectomy for whom and at what age? Primum non nocere. Maturitas. 2012, 71, 1-2.
    Ramos-Pratts K, Rosa-González D, Pérez-Acevedo NL, Cintrón-López D, Barreto-Estrada JL. Sex-specific effect of the anabolic steroid, 17α-methyltestosterone, on inhibitory avoidance learning in periadolescent rats. Behav Process. 2013, 99, 73-80.
    Reynolds RM. Glucocorticoid excess and the developmental origins of disease: two decades of testing the hypothesis--2012 Curt Richter Award Winner. Psychoneuroendocrinology. 2013, 38, 1-11.
    Stein-Behrens BA, Elliott EM, Miller CA, Schilling JW, Newcombe R, Sapolsky RM. Glucocorticoids exacerbate kainic acid-induced extracellular accumulation of excitatory amino acids in the rat hippocampus. J Neurochem. 1992, 58,1730-1735.
    Schultz R, Isola J, Parvinen M, Honkaniemi J, Wikström AC, Gustafsson JA, et al. Localization of the glucocorticoid receptor in testis and accessory sexual organs of male rat. Mol Cell Endocrinol. 1993, 95, 115-120.
    Shinwell ES, Karplus M, Reich D, Weintraub Z, Blazer S, Bader D, et al., Early postnatal dexamethasone treatment and increased incidence of cerebral palsy. Archives of Disease in Childhood-Fetal and Neonatal Edition. 2000, 83, F177-F181.
    Solum DT, Handa RJ. Estrogen regulates the development of brain-derived neurotrophic factor mRNA and protein in the rat hippocampus. J Neurosci. 2002, 22, 2650-2659.
    Stoffel EC, Craft RM. Ovarian hormone withdrawal-induced "depression" in female rats. Physiol Behav. 2004, 83, 505-513.
    Squire LR. Memory systems of the brain: a brief history and current perspective. Neurobiol Learn Mem. 2004, 82, 171-177.
    Suzuki S, Handa RJ. Regulation of estrogen receptor-beta expression in the female rat hypothalamus: differential effects of dexamethasone and estradiol. Endocrinology. 2004, 145, 3658-3670.
    Sherwin BB. Surgical menopause, estrogen, and cognitive function in women: what do the findings tell us? Ann NY Acad Sci. 2005, 1052, 3-10.
    Seckl JR. Glucocorticoids, developmental ‘programming’ and the risk of affective dysfunction. Prog Brain Res. 2008, 167, 17-34.
    Shappell NW, Hyndman KM, Bartell SE, Schoenfuss HL. Comparative biological effects and potency of 17α- and 17β-estradiol in fathead minnows. Aquat Toxicol. 2010, 100, 1-8.
    Sterrenburg L, Gaszner B, Boerrigter J, Santbergen L, Bramini M, Roubos EW, Peeters BW, Kozicz T. Sex-dependent and differential responses to acute restraint stress of corticotropin-releasing factor producing neurons in the rat paraventricular nucleus, central amygdala, and bed nucleus of the stria terminalis. J Neurosci Res. 2012, 90, 179-192.
    Struber N, Struber D, Roth G. Impact of early adversity on glucocorticoid regulation and later mental disorders. Neurosci Biobehav Rev 2014, 38, 17-37.
    Spies CM, Straub RH, Cutolo M, Buttgereit F. Circadian rhythms in rheumatology-a glucocorticoid perspective. Arthritis Res Ther. 2014, 16, S3.
    Singh P, Thakur MK. Histone deacetylase 2 inhibition attenuates downregulation of hippocampal plasticity gene expression during aging. Mol Neurobiol. 2018, 55,2432-2442.
    Terakawa N, Shimizu I, Aono T, Tanizawa O, Matsumoto K. Dexamethasone suppresses estrogen action at the pituitary level without modulating estrogen receptor dynamics. J Steroid Biochem. 1985, 23, 385-388.
    Toran-Allerand CD. Minireview: A plethora of estrogen receptors in the brain: where will it end? Endocrinology. 2004, 145, 1069-1074.
    Tonya White, Kathryn Cullen, Lisa Michelle Rohrer, Canan Karatekin, Monica Luciana, Marcus Schmidt, Donaya Hongwanishkul, Sanjiv Kumra, S. Charles Schulz, Kelvin O. Limbic Structures and Networks in Children and Adolescents With Schizophrenia. Schizophr Bull. 2008, 34, 18-29.
    Tiwari-Woodruff S, Voskuhl RR. Neuroprotective and anti-inflammatory effects of estrogen receptor ligand treatment in mice. J Neurol Sci. 2009, 286, 81-85.
    Toufexis D, Rivarola MA, Lara H, Viau V. Stress and the reproductive axis. J Neuroendocrinol. 2014, 26, 573-586.
    Ulrich-Lai YM, Herman JP. Neural regulation of endocrine and autonomic stress responses. Nat Rev Neurosci. 2009, 10, 397-409.
    Vandenbergh JG, Whitsett JM, Lombardi JR. Partial isolation of a pheromone accelerating puberty in female mice. J Reprod Fertil. 1975, 43, 515-523.
    van der Heide-Jalving M, Kamphuis PJ, van der Laan MJ, Bakker JM, Wiegant VM, Heijnen CJ, Veen S, van Bel F. Short- and long-term effects of neonatal glucocorticoid therapy: is hydrocortisone an alternative to dexamethasone? Acta Paediatr. 2003, 92, 827-835.
    Vasudevan N, Kow LM, Pfaff D. Integration of steroid hormone initiated membrane action to genomic function in the brain. Steroids. 2005, 70, 388-396.
    Verhaeghe J, Vanstapel F, Van Bree R, Van Herck E, Coopmans W. Transient catabolic state with reduced IGF-I after antenatal glucocorticoids. Pediatr Res. 2007, 62, 295-300.
    Woolley CS, Weiland NG, McEwen BS, Schwartzkroin PA. Estradiol increases the sensitivity of hippocampal CA1 pyramidal cells to NMDA receptor-mediated synaptic input: correlation with dendritic spine density. J Neurosci. 1997, 17, 1848-1859.
    Walf AA, Rhodes ME, Frye CA. Antidepressant effects of ER selective estrogen receptor modulators in the forced swim test. Pharmacol Biochem Be. 2004, 78, 523-529.
    Walf AA, Frye CA. A review and update of mechanisms of estrogen in the hippocampus and amygdala for anxiety and depression behavior. Neuropsychopharmacol. 2006, 31, 1097-1111.
    Weinstock M. The long-term behavioural consequences of prenatal stress. Neurosci Biobehav Rev. 2008, 32, 1073-1086.
    Wilson-Costello D, Walsh MC, Langer JC, Guillet R, Laptook AR, Stoll BJ, Shankaran S, Finer NN, Van Meurs KP, Engle WA, Das A; Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Das A Impact of postnatal corticosteroid use on neurodevelopment at 18 to 22 months' adjusted age: effects of dose, timing, and risk of bronchopulmonary dysplasia in extremely low birth weight infants. Pediatrics. 2009, 123, e430-e437.
    Whirledge S, Cidlowski JA. Glucocorticoids, stress, and fertility. Minerva Endocrinol. 2010, 35, 109-125.
    Walf AA, Frye CA. Estradiol reduces anxiety- and depression-like behavior of aged female mice. Physiol Behav. 2010, 99, 169-174.
    Waffarn F, Davis EP. Effects of antenatal corticosteroids on the hypothalamic-pituitary-adrenocortical axis of the fetus and newborn: experimental findings and clinical considerations. Am J Obstet Gynecol. 2012, 207, 446-454.
    Whirledge S, Cidlowski JA. A role for glucocorticoids in stress-impaired reproduction: beyond the hypothalamus and pituitary. Endocrinology. 2013, 154, 4450-4468.
    Yeh TF, Lin YJ, Hsieh WS, Lin HC, Lin CH, Chen JY, Kao HA, Chien CH. Early postnatal dexamethasone therapy for the prevention of chronic lung disease in preterm infants with respiratory distress syndrome: a multicenter clinical trial. Pediatrics. 1997, 100, E3.
    Yang X, Phillips DL, Ferguson AT, Nelson WG, Herman JG, Davidson NE. Synergistic activation of functional estrogen receptor (ER)-alpha by DNA methyltransferase and histone deacetylase inhibition in human ER-alpha-negative breast cancer cells. Cancer Res. 2001, 61, 7025-7029.
    Youngson NA, Whitelaw E. Transgenerational epigenetic effects. Annu Rev Genomics Hum Genet. 2008, 9, 233-257.
    Yang Y, Wang JZ. From structure to behavior in basolateral amygdala-hippocampus circuits. Front Neural Circuits. 2017, 11, 86.
    Zhu BT, Conney AH. Functional role of estrogen metabolism in target cells: review and perspectives. Carcinogenesis. 1998, 19, 1-27.
    Zhang WN, Bast T, Feldon J. The ventral hippocampus and fear conditioning in rats: different anterograde amnesias of fear after infusion of N-methyl-D-aspartate or its noncompetitive antagonist MK-801 into the ventral hippocampus. Behav Brain Res. 2001, 126, 159-174.
    Zhou Q, Atadja P, Davidson NE. Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biol Ther.. 2007, 6, 64-69.
    Zen M, Canova M, Campana C, Bettio S, Nalotto L, Rampudda M, Ramonda R, Iaccarino L, Doria A. The kaleidoscope of glucorticoid effects on immune system. Autoimmun Rev. 2011, 10, 305-310.
    Zuloaga DG, Carbone DL, Hiroi R, Chong DL, Handa RJ. Dexamethasone induces apoptosis in the developing rat amygdala in an age-, region-, and sex-specific manner. Neuroscience. 2011, 199, 535-547.
    Zhu L, Li H, Tang J, Zhu J, Zhang Y. Hyperoxia arrests alveolar development through suppression of histone deacetylases in neonatal rats. Pediatr Pulm. 2012, 47, 264-274.
    Zuloaga DG, Carbone DL, Handa RJ. Prenatal dexamethasone selectively decreases calretinin expression in the adult female lateral amygdala. Neurosci Lett. 2012a, 521, 109-114.
    Zuloaga DG, Carbone DL, Quihuis A, Hiroi R, Chong DL, Handa RJ. Perinatal dexamethasone-induced alterations in apoptosis within the hippocampus and paraventricular nucleus of the hypothalamus are influenced by age and sex. J Neurosci Res. 2012b, 90, 1403-1412.

    無法下載圖示 本全文未授權公開
    QR CODE