全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Estrogen Signaling and the Aging Brain: Context-Dependent Considerations for Postmenopausal Hormone Therapy

DOI: 10.1155/2013/814690

Full-Text   Cite this paper   Add to My Lib

Abstract:

Recent clinical studies have spurred rigorous debate about the benefits of hormone therapy (HT) for postmenopausal women. Controversy first emerged based on a sharp increase in the risk of cardiovascular disease in participants of the Women’s Health Initiative (WHI) studies, suggesting that decades of empirical research in animal models was not necessarily applicable to humans. However, a reexamination of the data from the WHI studies suggests that the timing of HT might be a critical factor and that advanced age and/or length of estrogen deprivation might alter the body's ability to respond to estrogens. Dichotomous estrogenic effects are mediated primarily by the actions of two high-affinity estrogen receptors alpha and beta (ERα & ERβ). The expression of the ERs can be overlapping or distinct, dependent upon brain region, sex, age, and exposure to hormone, and, during the time of menopause, there may be changes in receptor expression profiles, post-translational modifications, and protein:protein interactions that could lead to a completely different environment for E2 to exert its effects. In this review, factors affecting estrogen-signaling processes will be discussed with particular attention paid to the expression and transcriptional actions of ERβ in brain regions that regulate cognition and affect. 1. Introduction According to the CDC (2008), the average lifespan for women in the USA is ~81 years of age. While the average lifespan has been steadily increasing over the past century (~48 years in 1900), the average age at which reproductive senescence, menopause, occurs has remained relatively constant between 45–55 years of age [1, 2]. Including the prepubescent years, this leaves women living about half of their lives without high levels of circulating ovarian hormones. The two primary ovarian hormones are 17β-estradiol (E2) and progesterone, both of which are required for female reproduction. Many positive anecdotal experiences are reported during times in the reproductive cycle when E2 is high, sparking further investigation into the role of E2 in various nonreproductive processes, including those pertaining to cognition and mood. The vast majority of basic science studies have described positive effects of E2 on cognitive processes at a molecular level, and, importantly, older postmenopausal females exhibit significant deficits when performing tasks that require the use of working memory, attentional processing, and executive function [3–8]. The natural aging process is coincident with menopause, which confounds studies attempting to

References

[1]  C. Bengtsson, O. Lindquist, and L. Redvall, “Is the menopausal age rapidly changing?” Maturitas, vol. 1, no. 3, pp. 159–164, 1979.
[2]  A. Singh, S. Kaur, and I. Walia, “A historical perspective on menopause and menopausal age,” Bulletin of the Indian Institute of History of Medicine (Hyderabad), vol. 32, no. 2, pp. 121–135, 2002.
[3]  P. Verhaeghen and J. Cerella, “Aging, executive control, and attention: a review of meta-analyses,” Neuroscience and Biobehavioral Reviews, vol. 26, no. 7, pp. 849–857, 2002.
[4]  T. E. Wroolie, H. A. Kenna, K. E. Williams et al., “Differences in verbal memory performance in postmenopausal women receiving hormone therapy: 17β-estradiol versus conjugated equine estrogens,” American Journal of Geriatric Psychiatry, vol. 19, no. 9, pp. 792–802, 2011.
[5]  B. B. Sherwin, “Estrogenic effects on memory in women,” Annals of the New York Academy of Sciences, vol. 743, pp. 213–230, 1994.
[6]  B. B. Sherwin, “Hormones, mood, and cognitive functioning in postmenopausal women,” Obstetrics and Gynecology, vol. 87, no. 2, supplement, pp. 20S–26S, 1996.
[7]  B. B. Sherwin, “Sex hormones and psychological functioning in postmenopausal women,” Experimental Gerontology, vol. 29, no. 3-4, pp. 423–430, 1994.
[8]  S. M. Phillips and B. B. Sherwin, “Effects of estrogen on memory function in surgically menopausal women,” Psychoneuroendocrinology, vol. 17, no. 5, pp. 485–495, 1992.
[9]  R. Lindsay, J. M. Aitken, and J. B. Anderson, “Long term prevention of postmenopausal osteoporosis by oestrogen. Evidence for an increased bone mass after delayed onset of oestrogen treatment,” The Lancet, vol. 1, no. 7968, pp. 1038–1040, 1976.
[10]  J. E. Rossouw, R. L. Prentice, J. E. Manson et al., “Postmenopausal hormone therapy and risk of cardiovascular disease by age and years since menopause,” The Journal of the American Medical Association, vol. 297, no. 13, pp. 1465–1477, 2007.
[11]  Q.-G. Zhang, D. Han, R.-M. Wang et al., “C terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-α and the critical period hypothesis of estrogen neuroprotection,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 35, pp. E617–E624, 2011.
[12]  W. Krezel, S. Dupont, A. Krust, P. Chambon, and P. F. Chapman, “Increased anxiety and synaptic plasticity in estrogen receptor β-deficient mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 21, pp. 12278–12282, 2001.
[13]  H. ?stlund, E. Keller, and Y. L. Hurd, “Estrogen receptor gene expression in relation to neuropsychiatric disorders,” Annals of the New York Academy of Sciences, vol. 1007, pp. 54–63, 2003.
[14]  S. A. Shumaker, C. Legault, S. R. Rapp et al., “Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial,” Journal of the American Medical Association, vol. 289, no. 20, pp. 2651–2662, 2003.
[15]  S. A. Shumaker, C. Legault, L. Kuller et al., “Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: women's Health Initiative Memory Study,” Journal of the American Medical Association, vol. 291, no. 24, pp. 2947–2958, 2004.
[16]  S. R. Rapp, M. A. Espeland, S. A. Shumaker et al., “Effect of estrogen plus progestin on global cognitive function in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial,” Journal of the American Medical Association, vol. 289, no. 20, pp. 2663–2672, 2003.
[17]  V. W. Henderson, K. S. Benke, R. C. Green, L. A. Cupples, and L. A. Farrer, “Postmenopausal hormone therapy and Alzheimer's disease risk: interaction with age,” Journal of Neurology, Neurosurgery and Psychiatry, vol. 76, no. 1, pp. 103–105, 2005.
[18]  E. Garbe and S. Suissa, “Hormone replacement therapy and acute coronary syndromes: methodological issues between randomized and observational studies,” Human Reproduction, vol. 19, no. 1, pp. 8–13, 2004.
[19]  B. B. Braden, A. N. Garcia, S. E. Mennenga et al., “Cognitive-impairing effects of medroxyprogesterone acetate in the rat: independent and interactive effects across time,” Psychopharmacology, vol. 218, no. 2, pp. 405–418, 2011.
[20]  B. R. Bhavnani, S.-P. Tam, and X. Lu, “Structure activity relationships and differential interactions and functional activity of various equine estrogens mediated via estrogen receptors (ERs) ERα and ERβ,” Endocrinology, vol. 149, no. 10, pp. 4857–4870, 2008.
[21]  E. Hogervorst and S. Bandelow, “Sex steroids to maintain cognitive function in women after the menopause: a meta-analyses of treatment trials,” Maturitas, vol. 66, no. 1, pp. 56–71, 2010.
[22]  D. J. Mangelsdorf, C. Thummel, M. Beato et al., “The nuclear receptor super-family: the second decade,” Cell, vol. 83, no. 6, pp. 835–839, 1995.
[23]  E. V. Jensen, T. Suzuki, T. Kawashima, W. E. Stumpf, P. W. Jungblut, and E. R. DeSombre, “A two-step mechanism for the interaction of estradiol with rat uterus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 59, no. 2, pp. 632–638, 1968.
[24]  F. Naftolin, T. L. Horvath, R. L. Jakab, C. Leranth, N. Harada, and J. Balthazart, “Aromatase immunoreactivity in axon terminals of the vertebrate brain: an immunocytochemical study on quail, rat, monkey and human tissues,” Neuroendocrinology, vol. 63, no. 2, pp. 149–155, 1996.
[25]  C. E. Roselli, S. E. Abdelgadir, O. K. R?nnekleiv, and S. A. Klosterman, “Anatomic distribution and regulation of aromatase gene expression in the rat brain,” Biology of Reproduction, vol. 58, no. 1, pp. 79–87, 1998.
[26]  J. Balthazart and G. F. Ball, “Is brain estradiol a hormone or a neurotransmitter?” Trends in Neurosciences, vol. 29, no. 5, pp. 241–249, 2006.
[27]  T. R. Pak, Y. S. Rao, S. A. Prins, and N. N. Mott, “An emerging role for microRNAs in sexually dimorphic neurobiological systems,” Pflügers Archiv, vol. 465, no. 5, pp. 655–667, 2013.
[28]  K. Yamagata, S. Fujiyama, S. Ito et al., “Maturation of microRNA is hormonally regulated by a nuclear receptor,” Molecular Cell, vol. 36, no. 2, pp. 340–347, 2009.
[29]  Y. Masuhiro, Y. Mezaki, M. Sakari et al., “Splicing potentiation by growth factor signals via estrogen receptor phosphorylation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 23, pp. 8126–8131, 2005.
[30]  A. Tremblay, G. B. Tremblay, F. Labrie, and V. Giguère, “Ligand-Independent recruitment of SRC-1 to estrogen receptor β through phosphorylation of activation function AF-1,” Molecular Cell, vol. 3, no. 4, pp. 513–519, 1999.
[31]  S. H. Meijsing, M. A. Pufall, A. Y. So, D. L. Bates, L. Chen, and K. R. Yamamoto, “DNA binding site sequence directs glucocorticoid receptor structure and activity,” Science, vol. 324, no. 5925, pp. 407–410, 2009.
[32]  W. Bourguet, P. Germain, and H. Gronemeyer, “Nuclear receptor ligand-binding domains: three-dimensional structures, molecular interactions and pharmacological implications,” Trends in Pharmacological Sciences, vol. 21, no. 10, pp. 381–388, 2000.
[33]  A. Koide, C. Zhao, M. Naganuma et al., “Identification of regions within the F domain of the human estrogen receptor α that are important for modulating transactivation and protein-protein interactions,” Molecular Endocrinology, vol. 21, no. 4, pp. 829–842, 2007.
[34]  D. F. Skafar and S. Koide, “Understanding the human estrogen receptor-alpha using targeted mutagenesis,” Molecular and Cellular Endocrinology, vol. 246, no. 1-2, pp. 83–90, 2006.
[35]  E. C. Chang, J. Frasor, B. Komm, and B. S. Katzenellenbogen, “Impact of estrogen receptor β on gene networks regulated by estrogen receptor α in breast cancer cells,” Endocrinology, vol. 147, no. 10, pp. 4831–4842, 2006.
[36]  X. Zhu, I. Leav, Y.-K. Leung et al., “Dynamic regulation of estrogen receptor-β expression by DNA methylation during prostate cancer development and metastasis,” American Journal of Pathology, vol. 164, no. 6, pp. 2003–2012, 2004.
[37]  D. N. Petersen, G. T. Tkalcevic, P. H. Koza-Taylor, T. G. Turi, and T. A. Brown, “Identification of estrogen receptor β2, a functional variant of estrogen receptor β expressed in normal rat tissues,” Endocrinology, vol. 139, no. 3, pp. 1082–1092, 1998.
[38]  L. A. Helguero, M. H. Faulds, J.-?. Gustafsson, and L.-A. Haldosén, “Estrogen receptors alfa (ERα) and beta (ERβ) differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11,” Oncogene, vol. 24, no. 44, pp. 6605–6616, 2005.
[39]  P. C. Kulakosky, M. A. McCarty, S. C. Jernigan, K. E. Risinger, and C. M. Klinge, “Response element sequence modulates estrogen receptor α and β affinity and activity,” Journal of Molecular Endocrinology, vol. 29, no. 1, pp. 137–152, 2002.
[40]  O. M. V. Grober, M. Mutarelli, G. Giurato et al., “Global analysis of estrogen receptor beta binding to breast cancer cell genome reveals an extensive interplay with estrogen receptor alpha for target gene regulation,” BMC Genomics, vol. 12, article 36, 2011.
[41]  O. I. Vivar, X. Zhao, E. F. Saunier et al., “Estrogen receptor β binds to and regulates three distinct classes of target genes,” Journal of Biological Chemistry, vol. 285, no. 29, pp. 22059–22066, 2010.
[42]  G. G. J. M. Kuiper, E. Enmark, M. Pelto-Huikko, S. Nilsson, and J.-?. Gustafsson, “Cloning of a novel estrogen receptor expressed in rat prostate and ovary,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 12, pp. 5925–5930, 1996.
[43]  G. G. J. M. Kuiper, B. Carlsson, K. Grandien et al., “Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors and α and β,” Endocrinology, vol. 138, no. 3, pp. 863–870, 1997.
[44]  D. B. Dubal, H. Zhu, J. Yu et al., “Estrogen receptor alpha, not beta, is a critical link in estradiol-mediated protection against brain injury,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 4, pp. 1952–1957, 2001.
[45]  P. J. Shughrue, P. J. Scrimo, and I. Merchenthaler, “Evidence for the colocalization of estrogen receptor-β mRNA and estrogen receptor-α immunoreactivity in neurons of the rat forebrain,” Endocrinology, vol. 139, no. 12, pp. 5267–5270, 1998.
[46]  P. J. Shughrue, M. V. Lane, and I. Merchenthaler, “Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system,” Journal of Comparative Neurology, vol. 388, no. 4, pp. 507–525, 1997.
[47]  T. A. Milner, B. S. McEwen, S. Hayashi, C. J. Li, L. P. Reagan, and S. E. Alves, “Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites,” The Journal of Comparative Neurology, vol. 429, no. 3, pp. 355–371, 2001.
[48]  T. A. Milner, L. S. Lubbers, S. E. Alves, and B. S. McEwen, “Nuclear and extranuclear estrogen binding sites in the rat forebrain and autonomic medullary areas,” Endocrinology, vol. 149, no. 7, pp. 3306–3312, 2008.
[49]  T. A. Milner, K. Ayoola, C. T. Drake et al., “Ultrastructural localization of estrogen receptor β immunoreactivity in the rat hippocampal formation,” Journal of Comparative Neurology, vol. 491, no. 2, pp. 81–95, 2005.
[50]  A. E. Herbison and D. T. Theodosis, “Absence of estrogen receptor immunoreactivity in somatostatin (SRIF) neurons of the periventricular nucleus but sexually dimorphic colocalization of estrogen receptor and SRIF immunoreactivities in neurons of the bed nucleus of the stria terminalis,” Endocrinology, vol. 132, no. 4, pp. 1707–1714, 1993.
[51]  K. Kalita, S. Szymczak, and L. Kaczmarek, “Non-nuclear estrogen receptor β and α in the hippocampus of male and female rats,” Hippocampus, vol. 15, no. 3, pp. 404–412, 2005.
[52]  T. Ivanova and C. Beyer, “Ontogenetic expression and sex differences of aromatase and estrogen receptor-α/β mRNA in the mouse hippocampus,” Cell and Tissue Research, vol. 300, no. 2, pp. 231–237, 2000.
[53]  C. Y. Pau, K.-Y. F. Pau, and H. G. Spies, “Putative estrogen receptor β and α mRNA expression in male and female rhesus macaques,” Molecular and Cellular Endocrinology, vol. 146, no. 1-2, pp. 59–68, 1998.
[54]  A. L. Wijayaratne and D. P. McDonnell, “The human estrogen receptor-α is a ubiquitinated protein whose stability is affected differentially by agonists, antagonists, and selective estrogen receptor modulators,” Journal of Biological Chemistry, vol. 276, no. 38, pp. 35684–35692, 2001.
[55]  V. Bartella, P. Rizza, I. Barone et al., “Estrogen receptor beta binds Sp1 and recruits a corepressor complex to the estrogen receptor alpha gene promoter,” Breast Cancer Research and Treatment, vol. 134, no. 2, pp. 569–581, 2012.
[56]  L. C. Li, C. C. Yeh, D. Nojima, and R. Dahiya, “Cloning and characterization of human estrogen receptor beta promoter,” Biochemical and Biophysical Research Communications, vol. 275, no. 2, pp. 682–689, 2000.
[57]  E. A. Vladusic, A. E. Hornby, F. K. Guerra-Vladusic, J. Lakins, and R. Lupu, “Expression and regulation of estrogen receptor ? in human breast tumors and cell lines,” Oncology Reports, vol. 7, no. 1, pp. 157–167, 2000.
[58]  T. Hatsumi and Y. Yamamuro, “Downregulation of estrogen receptor gene expression by exogenous 17β-estradiol in the mammary glands of lactating mice,” Experimental Biology and Medicine, vol. 231, no. 3, pp. 311–316, 2006.
[59]  H. B. Patisaul, P. L. Whitten, and L. J. Young, “Regulation of estrogen receptor beta mRNA in the brain: opposite effects of 17β-estradiol and the phytoestrogen, coumestrol,” Molecular Brain Research, vol. 67, no. 1, pp. 165–171, 1999.
[60]  T. J. Brown, N. J. MacLusky, M. Shanabrough, and F. Naftolin, “Comparison of age- and sex-related changes in cell nuclear estrogen-binding capacity and progestin receptor induction in the rat brain,” Endocrinology, vol. 126, no. 6, pp. 2965–2972, 1990.
[61]  B. S. Rubin, T. O. Fox, and R. S. Bridges, “Estrogen binding in nuclear and cytosolic extracts from brain and pituitary of middle-aged female rats,” Brain Research, vol. 383, no. 1-2, pp. 60–67, 1986.
[62]  T. Funabashi, S. P. Kleopoulos, P. J. Brooks et al., “Changes in estrogenic regulation of estrogen receptor α mRNA and progesterone receptor mRNA in the female rat hypothalamus during aging: an in situ hybridization study,” Neuroscience Research, vol. 38, no. 1, pp. 85–92, 2000.
[63]  M. E. Wilson, K. L. Rosewell, M. L. Kashon, P. J. Shughrue, I. Merchenthaler, and P. M. Wise, “Age differentially influences estrogen receptor-α (ERα) and estrogen receptor-β (ERβ) gene expression in specific regions of the rat brain,” Mechanisms of Ageing and Development, vol. 123, no. 6, pp. 593–601, 2002.
[64]  W. S. Post, P. J. Goldschmidt-Clermont, C. C. Wilhide et al., “Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system,” Cardiovascular Research, vol. 43, no. 4, pp. 985–991, 1999.
[65]  J.-P. J. Issa, Y. L. Ottaviano, P. Celano, S. R. Hamilton, N. E. Davidson, and S. B. Baylin, “Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon,” Nature Genetics, vol. 7, no. 4, pp. 536–540, 1994.
[66]  T. Funabashi and F. Kimura, “Effects of estrogen and estrogen receptor messenger RNA levels in young and middle-aged female rats: comparison of medial preoptic area and mediobasal hypothalamus,” Acta Biologica Hungarica, vol. 45, no. 2–4, pp. 223–231, 1994.
[67]  M. A. Miller, P. E. Kolb, B. Planas, and M. A. Raskind, “Estrogen receptor and neurotensin/neuromedin-N gene expression in the preoptic area are unaltered with age in Fischer 344 female rats,” Endocrinology, vol. 135, no. 5, pp. 1986–1995, 1994.
[68]  T. R. Chakraborty, P. R. Hof, L. Ng, and A. C. Gore, “Stereologic analysis of estrogen receptor alpha (ER alpha) expression in rat hypothalamus and its regulation by aging and estrogen,” Journal of Comparative Neurology, vol. 466, no. 3, pp. 409–421, 2003.
[69]  J. M. Westberry, A. L. Trout, and M. E. Wilson, “Epigenetic regulation of estrogen receptor beta expression in the rat cortex during aging,” NeuroReport, vol. 22, no. 9, pp. 428–432, 2011.
[70]  T. R. Chakraborty, L. Ng, and A. C. Gore, “Age-related changes in estrogen receptor β in rat hypothalamus: a quantitative analysis,” Endocrinology, vol. 144, no. 9, pp. 4164–4171, 2003.
[71]  I. Poola, S. Koduri, S. Chatra, and R. Clarke, “Identification of twenty alternatively spliced estrogen receptor alpha mRNAs in breast cancer cell lines and tumors using splice targeted primer approach,” Journal of Steroid Biochemistry and Molecular Biology, vol. 72, no. 5, pp. 249–258, 2000.
[72]  T. A. Ishunina and D. F. Swaab, “Hippocampal estrogen receptor-alpha splice variant TADDI in the human brain in aging and Alzheimer's disease,” Neuroendocrinology, vol. 89, no. 2, pp. 187–199, 2009.
[73]  T. A. Ishunina and D. F. Swaab, “Estrogen receptor-α splice variants in the human brain,” Gynecological Endocrinology, vol. 24, no. 2, pp. 93–98, 2008.
[74]  T. A. Ishunina, F. P. M. Kruijver, R. Balesar, and D. F. Swaab, “Differential expression of estrogen receptor α and β immunoreactivity in the human supraoptic nucleus in relation to sex and aging,” Journal of Clinical Endocrinology and Metabolism, vol. 85, no. 9, pp. 3283–3291, 2000.
[75]  T. A. Ishunina, D. F. Fischer, and D. F. Swaab, “Estrogen receptor α and its splice variants in the hippocampus in aging and Alzheimer's disease,” Neurobiology of Aging, vol. 28, no. 11, pp. 1670–1681, 2007.
[76]  J. R. Tollervey, Z. Wang, T. Hortobágyi et al., “Analysis of alternative splicing associated with aging and neurodegeneration in the human brain,” Genome Research, vol. 21, no. 10, pp. 1572–1582, 2011.
[77]  N. N. Mott and T. R. Pak, “Characterisation of human oestrogen receptor beta (ERβ) splice variants in neuronal cells,” Journal of Neuroendocrinology, vol. 24, no. 10, pp. 1311–1321, 2012.
[78]  S. Inoue, S.-J. Hoshino, H. Miyoshi et al., “Identification of a ovel isoform of estrogen receptor, a potential inhibitor of estrogen action, in vascular smooth muscle cells,” Biochemical and Biophysical Research Communications, vol. 219, no. 3, pp. 766–772, 1996.
[79]  J. K. Skipper, L. J. Young, J. M. Bergeron, M. T. Tetzlaff, C. T. Osborn, and D. Crews, “Identification of an isoform of the estrogen receptor messenger RNA lacking exon four and present in the brain,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 15, pp. 7172–7175, 1993.
[80]  R. H. Price Jr., N. Lorenzon, and R. J. Handa, “Differential expression of estrogen receptor beta splice variants in rat brain: identification and characterization of a novel variant missing exon 4,” Brain Research. Molecular Brain Research, vol. 80, no. 2, pp. 260–268, 2000.
[81]  S. Chu and P. J. Fuller, “Identification of a splice variant of the rat estrogen receptor β gene,” Molecular and Cellular Endocrinology, vol. 132, no. 1-2, pp. 195–199, 1997.
[82]  B. Lu, E. Leygue, H. Dotzlaw, L. J. Murphy, L. C. Murphy, and P. H. Watson, “Estrogen receptor-β mRNA variants in human and murine tissues,” Molecular and Cellular Endocrinology, vol. 138, no. 1-2, pp. 199–203, 1998.
[83]  T. R. Pak, W. C. J. Chung, J. L. Roberts, and R. J. Handa, “Ligand-independent effects of estrogen receptor β on mouse gonadotropin-releasing hormone promoter activity,” Endocrinology, vol. 147, no. 4, pp. 1924–1931, 2006.
[84]  T. R. Pak, W. C. J. Chung, L. R. Hinds, and R. J. Handa, “Estrogen receptor-β mediates dihydrotestosterone-induced stimulation of the arginine vasopressin promoter in neuronal cells,” Endocrinology, vol. 148, no. 7, pp. 3371–3382, 2007.
[85]  Y. K. Leung, P. Mak, S. Hassan, and S. M. Ho, “Estrogen receptor (ER)-β isoforms: a key to understanding ER-β signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 35, pp. 13162–13167, 2006.
[86]  J. M. Wang, X. Hou, S. Adeosun et al., “A dominant negative ERβ splice variant determines the effectiveness of early or late estrogen therapy after ovariectomy in rats,” PLoS One, vol. 7, no. 3, Article ID e33493, 2012.
[87]  W. C. J. Chung, T. R. Pak, S. Suzuki, W. A. Pouliot, M. E. Andersen, and R. J. Handa, “Detection and localization of an estrogen receptor beta splice variant protein (ERβ2) in the adult female rat forebrain and midbrain regions,” Journal of Comparative Neurology, vol. 505, no. 3, pp. 249–267, 2007.
[88]  R. H. Price Jr., C. A. Butler, P. Webb, R. Uht, P. Kushner, and R. J. Handa, “A splice variant of estrogen receptor β missing exon 3 displays altered subnuclear localization and capacity for transcriptional activation,” Endocrinology, vol. 142, no. 5, pp. 2039–2049, 2001.
[89]  Y. Wang and R. J. Miksicek, “Identification of a dominant negative form of the human estrogen receptor,” Molecular Endocrinology, vol. 5, no. 11, pp. 1707–1715, 1991.
[90]  E. Küppers and C. Beyer, “Expression of estrogen receptor-α and β mRNA in the developing and adult mouse striatum,” Neuroscience Letters, vol. 276, no. 2, pp. 95–98, 1999.
[91]  C. Gundlah, S. G. Kohama, S. J. Mirkes, V. T. Garyfallou, H. F. Urbanski, and C. L. Bethea, “Distribution of estrogen receptor beta (ERβ) mRNA in hypothalamus, midbrain and temporal lobe of spayed macaque: continued expression with hormone replacement,” Brain Research. Molecular Brain Research, vol. 76, no. 2, pp. 191–204, 2000.
[92]  Y. Morishima, P. J. M. Murphy, D.-P. Li, E. R. Sanchez, and W. B. Pratt, “Stepwise assembly of a glucocorticoid receptor·hsp90 heterocomplex resolves two sequential ATP-dependent events involving first hsp70 and then hsp90 in opening of the steroid binding pocket,” Journal of Biological Chemistry, vol. 275, no. 24, pp. 18054–18060, 2000.
[93]  K. D. Dittmar and W. B. Pratt, “Folding of the glucocorticoid receptor by the reconstituted hsp90-based chaperone machinery. The initial hsp90·p60·hsp70-dependent step is sufficient for creating the steroid binding conformation,” Journal of Biological Chemistry, vol. 272, no. 20, pp. 13047–13054, 1997.
[94]  K. I. Kang, X. Meng, J. Devin-Leclerc et al., “The molecular chaperone Hsp90 can negatively regulate the activity of a glucocorticosteroid-dependent promoter,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 4, pp. 1439–1444, 1999.
[95]  K. Unno, H. Asakura, Y. Shibuya, M. Kaiho, S. Okada, and N. Oku Naoto, “Increase in basal level of Hsp70, consisting chiefly of constitutively expressed hsp70 (Hsc70) in aged rat brain,” Journals of Gerontology. Series A, vol. 55, no. 7, pp. B329–B335, 2000.
[96]  U. E. Olazabal, D. W. Pfaff, and C. V. Mobbs, “Sex differences in the regulation of heat shock protein 70 kDa and 90 kDa in the rat ventromedial hypothalamus by estrogen,” Brain Research, vol. 596, no. 1-2, pp. 311–314, 1992.
[97]  M. A. Pahlavani, M. D. Harris, S. A. Moore, and A. Richardson, “Expression of heat shock protein 70 in rat spleen lymphocytes is affected by age but not by food restriction,” Journal of Nutrition, vol. 126, no. 9, pp. 2069–2075, 1996.
[98]  A. R. Heydari, B. Wu, R. Takahashi, R. Strong, and A. Richardson, “Expression of heat shock protein 70 is altered by age and diet at the level of transcription,” Molecular and Cellular Biology, vol. 13, no. 5, pp. 2909–2918, 1993.
[99]  M. Sabbah, K.-I. I. Kang, L. Tora, and G. Redeuilh, “Oestrogen receptor facilitates the formation of preinitiation complex assembly: involvement of the general transcription factor TFIIB,” Biochemical Journal, vol. 336, part 3, pp. 639–646, 1998.
[100]  S.-Y. Wu, M. C. Thomas, S. Y. Hou, V. Likhite, and C.-M. Chiang, “Isolation of mouse TFIID and functional characterization of TBP and TFIID in mediating estrogen receptor and chromatin transcription,” Journal of Biological Chemistry, vol. 274, no. 33, pp. 23480–23490, 1999.
[101]  S. Ghosh and M. K. Thakur, “Tissue-specific expression of receptor-interacting protein in aging mouse,” Age, vol. 30, no. 4, pp. 237–243, 2008.
[102]  J. Frasor, J. M. Danes, B. Komm, K. C. N. Chang, C. Richard Lyttle, and B. S. Katzenellenbogen, “Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype,” Endocrinology, vol. 144, no. 10, pp. 4562–4574, 2003.
[103]  J. Frasor, J. M. Danes, C. C. Funk, and B. S. Katzenellenbogen, “Estrogen down-regulation of the corepressor N-CoR: mechanism and implications for estrogen derepression of N-CoR-regulated genes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 37, pp. 13153–13157, 2005.
[104]  N. J. McKenna and B. W. O'Malley, “Combinatorial control of gene expression by nuclear receptors and coregulators,” Cell, vol. 108, no. 4, pp. 465–474, 2002.
[105]  T.-P. Yao, G. Ku, N. Zhou, R. Scully, and D. M. Livingston, “The nuclear hormone receptor coactivator SRC-1 is a specific target of p300,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 20, pp. 10626–10631, 1996.
[106]  B. Hanstein, R. Eckner, J. DiRenzo et al., “p300 is a component of an estrogen receptor coactivator complex,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 21, pp. 11540–11545, 1996.
[107]  S. Halachmi, E. Marden, G. Martin, H. MacKay, C. Abbondanza, and M. Brown, “Estrogen receptor-associated proteins: possible mediators of hormone-induced transcriptions,” Science, vol. 264, no. 5164, pp. 1455–1458, 1994.
[108]  A. J. Horlein, A. M. Naar, T. Heinzel et al., “Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor,” Nature, vol. 377, no. 6548, pp. 397–404, 1995.
[109]  J. D. Chen and R. M. Evans, “A transcriptional co-repressor that interacts with nuclear hormone receptors,” Nature, vol. 377, no. 6548, pp. 454–457, 1995.
[110]  A. Malovannaya, R. B. Lanz, S. Y. Jung et al., “Analysis of the human endogenous coregulator complexome,” Cell, vol. 145, no. 5, pp. 787–799, 2011.
[111]  Z. Nawaz, D. M. Lonard, C. L. Smith et al., “The Angelman syndrome-associated protein, E6-AP, is a coactivator for the nuclear hormone receptor superfamily,” Molecular and Cellular Biology, vol. 19, no. 2, pp. 1182–1189, 1999.
[112]  B. Zheng, M. Han, M. Bernier, and J.-K. Wen, “Nuclear actin and actin-binding proteins in the regulation of transcription and gene expression,” FEBS Journal, vol. 276, no. 10, pp. 2669–2685, 2009.
[113]  W. A. Hofmann, L. Stojiljkovic, B. Fuchsova et al., “Actin is part of pre-initiation complexes and is necessary for transcription by RNA polymerase II,” Nature Cell Biology, vol. 6, no. 11, pp. 1094–1101, 2004.
[114]  K. Tokunaga, T. Shibuya, Y. Ishihama et al., “Nucleocytoplasmic transport of fluorescent mRNA in living mammalian cells: nuclear mRNA export is coupled to ongoing gene transcription,” Genes to Cells, vol. 11, no. 3, pp. 305–317, 2006.
[115]  R. Métivier, G. Penot, M. R. Hübner et al., “Estrogen receptor-α directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter,” Cell, vol. 115, no. 6, pp. 751–763, 2003.
[116]  C. Ambrosino, R. Tarallo, A. Bamundo et al., “Identification of a hormone-regulated dynamic nuclear actin network associated with estrogen receptor α in human breast cancer cell nuclei,” Molecular and Cellular Proteomics, vol. 9, no. 6, pp. 1352–1367, 2010.
[117]  F. Shao, R. Zhang, L. Dong, and K. Ying, “Overexpression of gelsolin-like actin-capping protein is associated with progression of lung adenocarcinoma,” Tohoku Journal of Experimental Medicine, vol. 225, no. 2, pp. 95–101, 2011.
[118]  K. Nishimura, H.-J. Ting, Y. Harada et al., “Modulation of androgen receptor transactivation by gelsolin: a newly identified androgen receptor coregulator,” Cancer Research, vol. 63, no. 16, pp. 4888–4894, 2003.
[119]  J. S. Ahn, I. S. Jang, D. I. Kim et al., “Aging-associated increase of gelsolin for apoptosis resistance,” Biochemical and Biophysical Research Communications, vol. 312, no. 4, pp. 1335–1341, 2003.
[120]  I. Nalvarte, T. Schwend, and J.-?. Gustafsson, “Proteomics analysis of the estrogen receptor α receptosome,” Molecular and Cellular Proteomics, vol. 9, no. 7, pp. 1411–1422, 2010.
[121]  L.-H. Miau, C.-J. Chang, B.-J. Shen, W.-H. Tsai, and S.-C. Lee, “Identification of heterogeneous nuclear ribonucleoprotein K (hnRNP K) as a repressor of C/EBPβ-mediated gene activation,” Journal of Biological Chemistry, vol. 273, no. 17, pp. 10784–10791, 1998.
[122]  A. Ostareck-Lederer, D. H. Ostareck, C. Cans et al., “c-Src-mediated phosphorylation of hnRNP K drives translational activation of specifically silenced mRNAs,” Molecular and Cellular Biology, vol. 22, no. 13, pp. 4535–4543, 2002.
[123]  P. S. Bagga, G. K. Arhin, and J. Wilusz, “DSEF-1 is a member of the hnRNP H family of RNA-binding proteins and stimulates pre-mRNA cleavage and polyadenylation in vitro,” Nucleic Acids Research, vol. 26, no. 23, pp. 5343–5350, 1998.
[124]  V. Markovtsov, J. M. Nikolic, J. A. Goldman, C. W. Turck, M.-Y. Chou, and D. L. Black, “Cooperative assembly of an hnRNP complex induced by a tissue-specific homolog of polypyrimidine tract binding protein,” Molecular and Cellular Biology, vol. 20, no. 20, pp. 7463–7479, 2000.
[125]  S. Y. Jung, A. Malovannaya, J. Wei, B. W. O'Malley, and J. Qin, “Proteomic analysis of steady-state nuclear hormone receptor coactivator complexes,” Molecular Endocrinology, vol. 19, no. 10, pp. 2451–2465, 2005.
[126]  W. Hong, R. J. Resnick, C. Rakowski, D. Shalloway, S. J. Taylor, and G. A. Blobel, “Physical and functional interaction between the transcriptional cofactor CBP and the KH domain protein Sam68,” Molecular Cancer Research, vol. 1, no. 1, pp. 48–55, 2002.
[127]  R. Shao, X. Wang, B. Weijdeg?rd et al., “Coordinate regulation of heterogeneous nuclear ribonucleoprotein dynamics by steroid hormones in the human fallopian tube and endometrium in vivo and in vitro,” American Journal of Physiology, vol. 302, no. 10, pp. E1269–E1282, 2012.
[128]  C. S. Woolley and B. S. McEwen, “Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat,” Journal of Neuroscience, vol. 12, no. 7, pp. 2549–2554, 1992.
[129]  C. S. Woolley, H. J. Wenzel, and P. A. Schwartzkroin, “Estradiol increases the frequency of multiple synapse boutons in the hippocampal CA1 region of the adult female rat,” The Journal of Comparative Neurology, vol. 373, no. 1, pp. 108–117, 1996.
[130]  C. S. Woolley, “Estrogen-mediated structural and functional synaptic plasticity in the female rat hippocampus,” Hormones and Behavior, vol. 34, no. 2, pp. 140–148, 1998.
[131]  D. P. Srivastava, K. M. Woolfrey, K. A. Jones et al., “Rapid enhancement of two-step wiring plasticity by estrogen and NMDA receptor activity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 38, pp. 14650–14655, 2008.
[132]  M. Ogiue-Ikeda, N. Tanabe, H. Mukai et al., “Rapid modulation of synaptic plasticity by estrogens as well as endocrine disrupters in hippocampal neurons,” Brain Research Reviews, vol. 57, no. 2, pp. 363–375, 2008.
[133]  N. J. Sandstrom and C. L. Williams, “Spatial memory retention is enhanced by acute and continuous estradiol replacement,” Hormones and Behavior, vol. 45, no. 2, pp. 128–135, 2004.
[134]  E. Hogervorst, J. Williams, M. Budge, W. Riedel, and J. Jolles, “The nature of the effect of female gonadal hormone replacement therapy on cognitive function in post-menopausal women: a meta-analysis,” Neuroscience, vol. 101, no. 3, pp. 485–512, 2000.
[135]  M. E. Bailey, A. C. J. Wang, J. Hao et al., “Interactive effects of age and estrogen on cortical neurons: implications for cognitive aging,” Neuroscience, vol. 191, pp. 148–158, 2011.
[136]  A. A. Walf, M. E. Rhodes, and C. A. Frye, “Ovarian steroids enhance object recognition in naturally cycling and ovariectomized, hormone-primed rats,” Neurobiology of Learning and Memory, vol. 86, no. 1, pp. 35–46, 2006.
[137]  V. N. Luine, L. F. Jacome, and N. J. Maclusky, “Rapid enhancement of visual and place memory by estrogens in rats,” Endocrinology, vol. 144, no. 7, pp. 2836–2844, 2003.
[138]  L. Fan, Z. Zhao, P. T. Orr, C. H. Chambers, M. C. Lewis, and K. M. Frick, “Estradiol-induced object memory consolidation in middle-aged female mice requires dorsal hippocampal extracellular signal-regulated kinase and phosphatidylinositol 3-kinase activation,” Journal of Neuroscience, vol. 30, no. 12, pp. 4390–4400, 2010.
[139]  D. B. Dubal and P. M. Wise, “Neuroprotective effects of estradiol in middle-aged female rats,” Endocrinology, vol. 142, no. 1, pp. 43–48, 2001.
[140]  S.-H. Yang, J. Shi, A. L. Day, and J. W. Simpkins, “Estradiol exerts neuroprotective effects when administered after ischemic insult,” Stroke, vol. 31, no. 3, pp. 745–750, 2000.
[141]  J. W. Simpkins, G. Rajakumar, Y.-Q. Zhang et al., “Estrogens may reduce mortality and ischemic damage caused by middle cerebral artery occlusion in the female rat,” Journal of Neurosurgery, vol. 87, no. 5, pp. 724–730, 1997.
[142]  J. Shi, K. S. Panickar, S.-H. Yang, O. Rabbani, A. L. Day, and J. W. Simpkins, “Estrogen attenuates over-expression of β-amyloid precursor protein messager RNA in an animal model of focal ischemia,” Brain Research, vol. 810, no. 1-2, pp. 87–92, 1998.
[143]  Q.-H. Zhang, Y.-H. Huang, Y.-Z. Hu et al., “Disruption of estrogen receptor beta in mice brain results in pathological alterations resembling Alzheimer disease,” Acta Pharmacologica Sinica, vol. 25, no. 4, pp. 452–457, 2004.
[144]  E. F. Rissman, A. L. Heck, J. E. Leonard, M. A. Shupnik, and J.-?. Gustafsson, “Disruption of estrogen receptor β gene impairs spatial learning in female mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 6, pp. 3996–4001, 2002.
[145]  M. Day, A. Sung, S. Logue, M. Bowlby, and R. Arias, “Beta estrogen receptor knockout (BERKO) mice present attenuated hippocampal CA1 long-term potentiation and related memory deficits in contextual fear conditioning,” Behavioural Brain Research, vol. 164, no. 1, pp. 128–131, 2005.
[146]  R. Vierk, G. Glassmeier, L. Zhou et al., “Aromatase inhibition abolishes LTP generation in female but not in male mice,” The Journal of Neuroscience, vol. 32, no. 24, pp. 8116–8126, 2012.
[147]  R.-M. Vouimba, M. R. Foy, J. G. Foy, and R. F. Thompson, “17β-estradiol suppresses expression of long-term depression in aged rats,” Brain Research Bulletin, vol. 53, no. 6, pp. 783–787, 2000.
[148]  C. E. Andreescu, B. A. Milojkovic, E. D. Haasdijk et al., “Estradiol improves cerebellar memory formation by activating estrogen receptor β,” Journal of Neuroscience, vol. 27, no. 40, pp. 10832–10839, 2007.
[149]  Y.-Y. Huang, P. V. Nguyen, T. Abel, and E. R. Kandel, “Long-lasting forms of synaptic potentiation in the mammalian hippocampus,” Learning Memory, vol. 3, no. 2-3, pp. 74–85, 1996.
[150]  H. Mukai, T. Tsurugizawa, G. Murakami et al., “Rapid modulation of long-term depression and spinogenesis via synaptic estrogen receptors in hippocampal principal neurons,” Journal of Neurochemistry, vol. 100, no. 4, pp. 950–967, 2007.
[151]  L. Liu, T. P. Wong, M. F. Pozza et al., “Role of NMDA receptor subtypes in governing the direction of hippocampal synaptic plasticity,” Science, vol. 304, no. 5673, pp. 1021–1024, 2004.
[152]  M. Cyr, O. Ghribi, C. Thibault, M. Morissette, M. Landry, and T. di Paolo, “Ovarian steroids and selective estrogen receptor modulators activity on rat brain NMDA and AMPA receptors,” Brain Research Reviews, vol. 37, no. 1–3, pp. 153–161, 2001.
[153]  M. M. Adams, S. E. Fink, W. G. M. Janssen, R. A. Shah, and J. H. Morrison, “Estrogen modulates synaptic N-methyl-D-aspartate receptor subunit distribution in the aged hippocampus,” Journal of Comparative Neurology, vol. 474, no. 3, pp. 419–426, 2004.
[154]  C. C. Smith and L. L. McMahon, “Estradiol-induced increase in the magnitude of long-term potentiation is prevented by blocking NR2B-containing receptors,” Journal of Neuroscience, vol. 26, no. 33, pp. 8517–8522, 2006.
[155]  Y. Zhou, J. J. Watters, and D. M. Dorsa, “Estrogen rapidly induces the phosphorylation of the cAMP response element binding protein in rat brain,” Endocrinology, vol. 137, no. 5, pp. 2163–2166, 1996.
[156]  X. Gonda, T. Telek, G. Juhász, J. Lazary, A. Vargha, and G. Bagdy, “Patterns of mood changes throughout the reproductive cycle in healthy women without premenstrual dysphoric disorders,” Progress in Neuro-Psychopharmacology and Biological Psychiatry, vol. 32, no. 8, pp. 1782–1788, 2008.
[157]  E. W. Freeman, “Premenstrual syndrome and premenstrual dysphoric disorder: definitions and diagnosis,” Psychoneuroendocrinology, vol. 28, supplement 3, pp. 25–37, 2003.
[158]  T. D. Lund, T. Rovis, W. C. J. Chung, and R. J. Handa, “Novel actions of estrogen receptor-β on anxiety-related behaviors,” Endocrinology, vol. 146, no. 2, pp. 797–807, 2005.
[159]  N. Breslau, L. Schultz, and E. Peterson, “Sex differences in depression: a role for preexisting anxiety,” Psychiatry Research, vol. 58, no. 1, pp. 1–12, 1995.
[160]  P. E. Bebbington, G. Dunn, R. Jenkins et al., “The influence of age and sex on the prevalence of depressive conditions: report from the National Survey of Psychiatric Morbidity,” Psychological Medicine, vol. 28, no. 1, pp. 9–19, 1998.
[161]  G. Aguilera, J. P. Harwood, and J. X. Wilson, “Mechanisms of action of corticotropin-releasing factor and other regulators of corticotropin release in rat pituitary cells,” Journal of Biological Chemistry, vol. 258, no. 13, pp. 8039–8045, 1983.
[162]  A. Papadimitriou and K. N. Priftis, “Regulation of the hypothalamic-pituitary-adrenal axis,” NeuroImmunoModulation, vol. 16, no. 5, pp. 265–271, 2009.
[163]  S. Suzuki and R. J. Handa, “Regulation of estrogen receptor-β expression in the female rat hypothalamus: differential effects of dexamethasone and estradiol,” Endocrinology, vol. 145, no. 8, pp. 3658–3670, 2004.
[164]  W. J. S. Miller, S. Suzuki, L. K. Miller, R. Handa, and R. M. Uht, “Estrogen receptor (ER)β isoforms rather than ERα regulate corticotropin-releasing hormone promoter activity through an alternate pathway,” Journal of Neuroscience, vol. 24, no. 47, pp. 10628–10635, 2004.
[165]  C. Isgor, M. Cecchi, M. Kabbaj, H. Akil, and S. J. Watson, “Estrogen receptor β in the paraventricular nucleus of hypothalamus regulates the neuroendocrine response to stress and is regulated by corticosterone,” Neuroscience, vol. 121, no. 4, pp. 837–845, 2003.
[166]  M. L. Forsling, I. Kalló, D. E. Hartley et al., “Oestrogen receptor-β and neurohypophysial hormones: functional interaction and neuroanatomical localisation,” Pharmacology Biochemistry and Behavior, vol. 76, no. 3-4, pp. 535–542, 2003.
[167]  A. S. Lalmansingh and R. M. Uht, “Estradiol regulates corticotropin-releasing hormone gene (crh) expression in a rapid and phasic manner that parallels estrogen receptor-α and -β recruitment to a 3′,5′-cyclic adenosine 5′- monophosphate regulatory region of the proximal crh promoter,” Endocrinology, vol. 149, no. 1, pp. 346–357, 2008.
[168]  B. N. Roy, R. L. Reid, and D. A. van Vugt, “The effects of estrogen and progesterone on corticotropin-releasing hormone and arginine vasopressin messenger ribonucleic acid levels in the paraventricular nucleus and supraoptic nucleus of the rhesus monkey,” Endocrinology, vol. 140, no. 5, pp. 2191–2198, 1999.
[169]  X. Ni, R. C. Nicholson, B. R. King, E.-C. Chan, M. A. Read, and R. Smith, “Estrogen represses whereas the estrogen-antagonist ICI 182780 stimulates placental CRH gene expression,” Journal of Clinical Endocrinology and Metabolism, vol. 87, no. 8, pp. 3774–3778, 2002.
[170]  N. C. Vamvakopoulos and G. P. Chrousos, “Evidence of direct estrogenic regulation of human corticotropin-releasing hormone gene expression. Potential implications for the sexual dimophism of the stress response and immune/inflammatory reaction,” Journal of Clinical Investigation, vol. 92, no. 4, pp. 1896–1902, 1993.
[171]  Y. Hu, D. L. Wu, C. X. Luo et al., “Hippocampal nitric oxide contributes to sex difference in affective behaviors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 35, pp. 14224–14229, 2012.
[172]  B. E. H. Sumner and G. Fink, “Estrogen increases the density of 5-hydroxytryptamine(2A) receptors in cerebral cortex and nucleus accumbens in the female rat,” Journal of Steroid Biochemistry and Molecular Biology, vol. 54, no. 1-2, pp. 15–20, 1995.
[173]  L. J. Smith, J. A. Henderson, C. W. Abell, and C. L. Bethea, “Effects of ovarian steroids and raloxifene on proteins that synthesize, transport, and degrade serotonin in the raphe region of macaques,” Neuropsychopharmacology, vol. 29, no. 11, pp. 2035–2045, 2004.
[174]  D. B. Imwalle, J.-?. Gustafsson, and E. F. Rissman, “Lack of functional estrogen receptor β influences anxiety behavior and serotonin content in female mice,” Physiology and Behavior, vol. 84, no. 1, pp. 157–163, 2005.
[175]  K. Tomihara, T. Soga, M. Nomura et al., “Effect of ER-β gene disruption on estrogenic regulation of anxiety in female mice,” Physiology and Behavior, vol. 96, no. 2, pp. 300–306, 2009.
[176]  A. A. Walf, C. Koonce, K. Manley, and C. A. Frye, “Proestrous compared to diestrous wildtype, but not estrogen receptor beta knockout, mice have better performance in the spontaneous alternation and object recognition tasks and reduced anxiety-like behavior in the elevated plus and mirror maze,” Behavioural Brain Research, vol. 196, no. 2, pp. 254–260, 2009.
[177]  A. A. Walf, C. J. Koonce, and C. A. Frye, “Estradiol or diarylpropionitrile administration to wild type, but not estrogen receptor beta knockout, mice enhances performance in the object recognition and object placement tasks,” Neurobiology of Learning and Memory, vol. 89, no. 4, pp. 513–521, 2008.
[178]  A. A. Walf, C. J. Koonce, and C. A. Frye, “Estradiol or diarylpropionitrile decrease anxiety-like behavior of wildtype, but not estrogen receptor beta knockout, mice,” Behavioral Neuroscience, vol. 122, no. 5, pp. 974–981, 2008.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413