Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The neuroprotective actions of oestradiol and oestrogen receptors

Key Points

  • Oestradiol is both a sex steroid hormone and a neurosteroid that is locally synthesized in the brain. Both hormonal oestradiol and brain-derived oestradiol are neuroprotective.

  • Oestradiol synthesis in the brain is rapidly regulated in neurons by synaptic activity. In turn, brain-derived oestradiol regulates synaptic plasticity, adult neurogenesis, reproductive behaviour, aggressive behaviour, pain processing, affect and cognition.

  • Under pathological conditions, the expression of aromatase, the enzyme that produces oestradiol, is enhanced in neurons and induced de novo in astrocytes as an endogenous neuroprotective mechanism. Inhibition or silencing of brain aromatase increases gliosis and neurodegeneration after brain injury.

  • The neuroprotective actions of oestradiol are mediated by two oestrogen receptors (ERs) located in the cell nucleus, ERα and ERβ, and by ERs located in the membrane, including ERα, ERβ, G protein-coupled ER and Gαq protein-coupled ER.

  • The ERs coordinate various neuroprotective signalling mechanisms, some of which are complementary and some of which are redundant. These include the regulation of transcription by nuclear ERs and the regulation of the activity of different kinases by membrane ERs.

  • Membrane and intracellular ERs also contribute to the interaction of oestradiol signalling with the signalling of other neuroprotective factors, such as brain-derived neurotrophic factor, insulin-like growth factor 1, WNT and Notch.

  • Further studies are necessary to determine the role of neuronal and non-neuronal cells in the coordination of oestradiol-mediated neuroprotective signalling mechanisms. The influence of sex and age on these mechanisms should also be studied.

  • A promising research direction aims to determine the role of metabolic homeostatic regulation in the protective actions of oestradiol, particularly in chronic neurodegenerative diseases.

  • Further research should also explore alternatives to oestradiol therapy, including new ligands for ERs and modulators of brain oestradiol synthesis.

Abstract

Hormones regulate homeostasis by communicating through the bloodstream to the body's organs, including the brain. As homeostatic regulators of brain function, some hormones exert neuroprotective actions. This is the case for the ovarian hormone 17β-oestradiol, which signals through oestrogen receptors (ERs) that are widely distributed in the male and female brain. Recent discoveries have shown that oestradiol is not only a reproductive hormone but also a brain-derived neuroprotective factor in males and females and that ERs coordinate multiple signalling mechanisms that protect the brain from neurodegenerative diseases, affective disorders and cognitive decline.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Neuroprotective actions of brain aromatase.
Figure 2: Oestradiol activates multiple neuroprotective signalling mechanisms.
Figure 3: Redundant neuroprotective signalling elicited by oestrogen receptors.
Figure 4: Oestradiol mediates indirect regulation of other neuroprotective signalling pathways.

Similar content being viewed by others

References

  1. Baulieu, E. E. Neurosteroids: a novel function of the brain. Psychoneuroendocrinology 23, 963–987 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Do Rego, J. L. et al. Neurosteroid biosynthesis: enzymatic pathways and neuroendocrine regulation by neurotransmitters and neuropeptides. Front. Neuroendocrinol. 30, 259–301 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Caruso, D. et al. Comparison of plasma and cerebrospinal fluid levels of neuroactive steroids with their brain, spinal cord and peripheral nerve levels in male and female rats. Psychoneuroendocrinology 38, 2278–2290 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Fokidis, H. B. et al. Regulation of local steroidogenesis in the brain and in prostate cancer: Lessons learned from interdisciplinary collaboration. Front. Neuroendocrinol. 16 Sep 2014 (10.1016/j.yfrne.2014.08.005).

  5. Caruso, D. et al. Effect of short-and long-term gonadectomy on neuroactive steroid levels in the central and peripheral nervous system of male and female rats. J. Neuroendocrinol. 22, 1137–1147 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Azcoitia, I., Fernandez-Galaz, C., Sierra, A. & Garcia-Segura, L. M. Gonadal hormones affect neuronal vulnerability to excitotoxin-induced degeneration. J. Neurocytol. 28, 699–710 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Azcoitia, I. et al. Brain aromatase is neuroprotective. J. Neurobiol. 47, 318–329 (2001). Shows that brain aromatase inhibition increases excitotoxic neuronal death in the hippocampus after kainic acid administration in male rats.

    Article  CAS  PubMed  Google Scholar 

  8. Yue, X. et al. Brain estrogen deficiency accelerates Aβ plaque formation in an Alzheimer's disease animal model. Proc. Natl Acad. Sci. USA 102, 19198–19203 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Overk, C. R. et al. Effects of aromatase inhibition versus gonadectomy on hippocampal complex amyloid pathology in triple transgenic mice. Neurobiol. Dis. 45, 479–487 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Ding, F. et al. Ovariectomy induces a shift in fuel availability and metabolism in the hippocampus of the female transgenic model of familial Alzheimer's. PLoS ONE 8, e59825 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Scott, E., Zhang, Q. G., Wang, R., Vadlamudi, R. & Brann, D. Estrogen neuroprotection and the critical period hypothesis. Front. Neuroendocrinol. 33, 85–104 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Tang, M. X. et al. Effect of oestrogen during menopause on risk and age at onset of Alzheimer's disease. Lancet 348, 429–432 (1996).

    Article  CAS  PubMed  Google Scholar 

  13. Sherwin, B. B. Estrogen and cognitive functioning in women. Endocr. Rev. 24, 133–151 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Garcia-Segura, L. M. et al. Aromatase expression by astrocytes after brain injury: implications for local estrogen formation in brain repair. Neuroscience 89, 567–578 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Peterson, R. S., Lee, D. W., Fernando, G. & Schlinger, B. A. Radial glia express aromatase in the injured zebra finch brain. J. Comp. Neurol. 475, 261–269 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Carswell, H. V. et al. Brain aromatase expression after experimental stroke: topography and time course. J. Steroid Biochem. Mol. Biol. 96, 89–91 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Zhang, Q. G. et al. Brain-derived estrogen exerts anti-inflammatory and neuroprotective actions in the rat hippocampus. Mol. Cell Endocrinol. 389, 84–91 (2014). Demonstrates that brain aromatase silencing enhances neuronal death and neuroinflammation in CA1 after global cerebral ischaemia in female rats.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gatson, J. W. et al. Aromatase is increased in astrocytes in the presence of elevated pressure. Endocrinology 152, 207–213 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Zuloaga, K. L., Davis, C. M., Zhang, W. & Alkayed, N. J. Role of aromatase in sex-specific cerebrovascular endothelial function in mice. Am. J. Physiol. Heart Circ. Physiol. 306, H929–H937 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. McCullough, L. D., Blizzard, K., Simpson, E. R., Oz, O. K. & Hurn, P. D. Aromatase cytochrome P450 and extragonadal estrogen play a role in ischemic neuroprotection. J. Neurosci. 23, 8701–8705 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sierra, A., Azcoitia, I. & Garcia-Segura, L. Endogenous estrogen formation is neuroprotective in model of cerebellar ataxia. Endocrine 21, 43–51 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Li, J. et al. Estrogen enhances neurogenesis and behavioral recovery after stroke. J. Cereb. Blood Flow Metab. 31, 413–425 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Wynne, R. D., Walters, B. J., Bailey, D. J. & Saldanha, C. J. Inhibition of injury-induced glial aromatase reveals a wave of secondary degeneration in the songbird brain. Glia 56, 97–105 (2008). Shows that inhibition of brain aromatase in songbirds increases gliosis and neuronal degeneration after brain injury.

    Article  PubMed  Google Scholar 

  24. Stanic, D. et al. Characterization of aromatase expression in the adult male and female mouse brain. I. Coexistence with oestrogen receptors α and β, and androgen receptors. PLoS ONE 9, e90451 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Iivonen, S. et al. Polymorphisms in the CYP19 gene confer increased risk for Alzheimer disease. Neurology 62, 1170–1176 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Huang, R. & Poduslo, S. E. CYP19 haplotypes increase risk for Alzheimer's disease. J. Med. Genet. 43, e42 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chace, C. et al. Variants in CYP17 and CYP19 cytochrome P450 genes are associated with onset of Alzheimer's disease in women with down syndrome. J. Alzheimers Dis. 28, 601–612 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Medway, C. et al. The sex-specific associations of the aromatase gene with Alzheimer's disease and its interaction with IL10 in the Epistasis Project. Eur. J. Hum. Genet. 22, 216–220 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Luchetti, S. et al. Neurosteroid biosynthetic pathways changes in prefrontal cortex in Alzheimer's disease. Neurobiol. Aging 32, 1964–1976 (2011). Presents an analysis of the expression of key steroidogenic enzymes in the prefrontal cortex of patients with Alzheimer's disease.

    Article  CAS  PubMed  Google Scholar 

  30. Green, S. et al. Human oestrogen receptor cDNA: sequence, expression and homology to v-erb-A. Nature 320, 134–139 (1986).

    Article  CAS  PubMed  Google Scholar 

  31. Greene, G. L. et al. Sequence and expression of human estrogen receptor complementary DNA. Science 231, 1150–1154 (1986).

    Article  CAS  PubMed  Google Scholar 

  32. Kuiper, G. G., Enmark, E., Pelto-Huikko, M., Nilsson, S. & Gustafsson, J. A. Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Natl Acad. Sci. USA 93, 5925–5930 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Shang, Y., Hu, X., DiRenzo, J., Lazar, M. A. & Brown, M. Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell 103, 843–852 (2000).

    Article  CAS  Google Scholar 

  34. Safe, S. & Kim, K. Non-classical genomic estrogen receptor (ER)/specificity protein and ER/activating protein-1 signaling pathways. J. Mol. Endocrinol. 41, 263–275 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ma, Z. Q. et al. Insulin-like growth factors activate estrogen receptor to control the growth and differentiation of the human neuroblastoma cell line SK-ER3. Mol. Endocrinol. 8, 910–918 (1994).

    CAS  PubMed  Google Scholar 

  36. Mendez, P. & Garcia-Segura, L. M. Phosphatidylinositol 3-kinase and glycogen synthase kinase 3 regulate estrogen receptor-mediated transcription in neuronal cells. Endocrinology 147, 3027–3039 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Ramirez, C. M. et al. VDAC and ERα interaction in caveolae from human cortex is altered in Alzheimer's disease. Mol. Cell Neurosci. 42, 172–183 (2009). Demonstrates localization of ERα in lipid rafts in the human brain, confirming previous studies in rodents.

    Article  CAS  PubMed  Google Scholar 

  38. Ruiz-Palmero, I., Hernando, M., Garcia-Segura, L. M. & Arevalo, M.-A. G protein-coupled estrogen receptor is required for the neuritogenic mechanism of 17β-estradiol in developing hippocampal neurons. Mol. Cell Endocrinol. 372, 105–115 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Qiu, J. et al. Rapid signaling of estrogen in hypothalamic neurons involves a novel G-protein-coupled estrogen receptor that activates protein kinase C. J. Neurosci. 23, 9529–9540 (2003). Details the identification of a new membrane ER (G q -mER) in hypothalamic neurons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Miller, N. R., Jover, T., Cohen, H. W., Zukin, R. S. & Etgen, A. M. Estrogen can act via estrogen receptor α and β to protect hippocampal neurons against global ischemia-induced cell death. Endocrinology 146, 3070–3079 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Zhao, L. & Brinton, R. D. Estrogen receptor α and β differentially regulate intracellular Ca2+ dynamics leading to ERK phosphorylation and estrogen neuroprotection in hippocampal neurons. Brain Res. 1172, 48–59 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Lebesgue, D. et al. Acute administration of non-classical estrogen receptor agonists attenuates ischemia-induced hippocampal neuron loss in middle-aged female rats. PLoS ONE 5, e8642 (2010). Shows that the non-classical ERs GPER and G q -mER participate in the neuroprotective actions of oestradiol when given after ischaemia.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Spence, R. D. et al. Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERα signaling on astrocytes but not through ERβ signaling on astrocytes or neurons. J. Neurosci. 33, 10924–10933 (2013). Together with a previous study by this group, this paper shows that, in an experimental model of multiple sclerosis, the protective action of ERα agonists is mediated by ERα expressed in astrocytes but not by ERα expressed in neurons and that the protective role of ERβ agonists is not mediated by ERβ expressed in astrocytes or neurons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Madinier, A., Wieloch, T., Olsson, R. & Ruscher, K. Impact of estrogen receptor β activation on functional recovery after experimental stroke. Behav. Brain Res. 261, 282–288 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Tang, H. et al. GPR30 mediates estrogen rapid signaling and neuroprotection. Mol. Cell Endocrinol. 387, 52–58 (2014). This paper reports that GPER is involved in the neuroprotective actions of oestradiol in the hippocampus after global cerebral ischaemia in adult female rats. GPER activates ERK and AKT signalling and reduces JNK signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dubal, D. B. et al. Differential modulation of estrogen receptors (ERs) in ischemic brain injury: a role for ERα in estradiol-mediated protection against delayed cell death. Endocrinology 147, 3076–3084 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Zhang, Q. G. et al. Estrogen attenuates ischemic oxidative damage via an estrogen receptor α-mediated inhibition of NADPH oxidase activation. J. Neurosci. 29, 13823–13836 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Simpkins, J. W., Singh, M., Brock, C. & Etgen, A. M. Neuroprotection and estrogen receptors. Neuroendocrinology 96, 119–130 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Dubal, D. B. et al. Estrogen receptor α, not β, is a critical link in estradiol-mediated protection against brain injury. Proc. Natl Acad. Sci. USA 98, 1952–1957 (2001).

    CAS  PubMed  Google Scholar 

  50. Suzuki, S. et al. Estradiol enhances neurogenesis following ischemic stroke through estrogen receptors α and β. J. Comp. Neurol. 500, 1064–1075 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Carswell, H. V., Macrae, I. M., Gallagher, L., Harrop, E. & Horsburgh, K. J. Neuroprotection by a selective estrogen receptor β agonist in a mouse model of global ischemia. Am. J. Physiol. Heart Circ. Physiol. 287, H1501–H1504 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Ciriza, I., Carrero, P., Azcoitia, I., Lundeen, S. G. & Garcia-Segura, L. M. Selective estrogen receptor modulators protect hippocampal neurons from kainic acid excitotoxicity: differences with the effect of estradiol. J. Neurobiol. 61, 209–221 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Morissette, M., Al Sweidi, S., Callier, S. & Di Paolo, T. Estrogen and SERM neuroprotection in animal models of Parkinson's disease. Mol. Cell Endocrinol. 290, 60–69 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. DonCarlos, L. L., Azcoitia, I. & Garcia-Segura, L. M. Neuroprotective actions of selective estrogen receptor modulators. Psychoneuroendocrinology 34, S113–S122 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Chowen, J. A., Torres-Aleman, I. & Garcia-Segura, L. M. Trophic effects of estradiol on fetal rat hypothalamic neurons. Neuroendocrinology 56, 895–901 (1992).

    Article  CAS  PubMed  Google Scholar 

  56. Valles, S. L. et al. Oestradiol or genistein rescues neurons from amyloid β-induced cell death by inhibiting activation of p38. Aging Cell 7, 112–118 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Gingerich, S. et al. Estrogen receptor α and G-protein coupled receptor 30 mediate the neuroprotective effects of 17β-estradiol in novel murine hippocampal cell models. Neuroscience 170, 54–66 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Cui, J. et al. Morphine protects against intracellular amyloid toxicity by inducing estradiol release and upregulation of Hsp70. J. Neurosci. 31, 16227–16240 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sierra, A. et al. Steroidogenic acute regulatory protein in the rat brain: cellular distribution, developmental regulation and overexpression after injury. Eur. J. Neurosci. 18, 1458–1467 (2003).

    Article  PubMed  Google Scholar 

  60. Schaeffer, V., Meyer, L., Patte-Mensah, C., Eckert, A. & Mensah-Nyagan, A. G. Sciatic nerve injury induces apoptosis of dorsal root ganglion satellite glial cells and selectively modifies neurosteroidogenesis in sensory neurons. Glia 58, 169–180 (2010).

    Article  PubMed  Google Scholar 

  61. Pietranera, L. et al. Increased aromatase expression in the hippocampus of spontaneously hypertensive rats: effects of estradiol administration. Neuroscience 174, 151–159 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Prange-Kiel, J., Wehrenberg, U., Jarry, H. & Rune, G. M. Para/autocrine regulation of estrogen receptors in hippocampal neurons. Hippocampus 13, 226–234 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Johann, S. & Beyer, C. Neuroprotection by gonadal steroid hormones in acute brain damage requires cooperation with astroglia and microglia. J. Steroid Biochem. Mol. Biol. 137, 71–81 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Dhandapani, K. M. & Brann, D. W. Role of astrocytes in estrogen-mediated neuroprotection. Exp. Gerontol. 42, 70–75 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Arevalo, M.-A., Santos-Galindo, M., Bellini, M. J., Azcoitia, I. & Garcia-Segura, L. M. Actions of estrogens on glial cells: Implications for neuroprotection. Biochim. Biophys. Acta 1800, 1106–1112 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Garcia-Ovejero, D., Azcoitia, I., Doncarlos, L. L., Melcangi, R. C. & Garcia-Segura, L. M. Glia-neuron crosstalk in the neuroprotective mechanisms of sex steroid hormones. Brain Res. Brain Res. Rev. 48, 273–286 (2005).

    Article  CAS  PubMed  Google Scholar 

  67. Acaz-Fonseca, E., Sanchez-Gonzalez, R., Azcoitia, I., Arevalo, M.-A. & Garcia-Segura, L. M. Role of astrocytes in the neuroprotective actions of 17β-estradiol and selective estrogen receptor modulators. Mol. Cell Endocrinol. 389, 48–57 (2014).

    Article  CAS  PubMed  Google Scholar 

  68. Garcia-Ovejero, D., Veiga, S., Garcia-Segura, L. M. & Doncarlos, L. L. Glial expression of estrogen and androgen receptors after rat brain injury. J. Comp. Neurol. 450, 256–271 (2002).

    Article  CAS  PubMed  Google Scholar 

  69. Sortino, M. A. et al. Glia mediates the neuroprotective action of estradiol on β-amyloid-induced neuronal death. Endocrinology 145, 5080–5086 (2004).

    Article  CAS  PubMed  Google Scholar 

  70. Dhandapani, K. M., Wade, F. M., Mahesh, V. B. & Brann, D. W. Astrocyte-derived transforming growth factor-β mediates the neuroprotective effects of 17β-estradiol: involvement of nonclassical genomic signaling pathways. Endocrinology 146, 2749–2759 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Vegeto, E. et al. Estrogen receptor-α mediates the brain antiinflammatory activity of estradiol. Proc. Natl Acad. Sci. USA 100, 9614–9619 (2003).

    Article  CAS  PubMed  Google Scholar 

  72. Liu, X. et al. Estrogen provides neuroprotection against activated microglia-induced dopaminergic neuronal injury through both estrogen receptor-α and estrogen receptor-β in microglia. J. Neurosci. Res. 81, 653–665 (2005).

    Article  CAS  PubMed  Google Scholar 

  73. Ghisletti, S., Meda, C., Maggi, A. & Vegeto, E. 17β-estradiol inhibits inflammatory gene expression by controlling NF-κB intracellular localization. Mol. Cell. Biol. 25, 2957–2968 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. De Marinis, E. et al. 17β-oestradiol anti-inflammatory effects in primary astrocytes require oestrogen receptor β-mediated neuroglobin up-regulation. J. Neuroendocrinol. 25, 260–270 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Yang, L. C. et al. Extranuclear estrogen receptors mediate the neuroprotective effects of estrogen in the rat hippocampus. PLoS ONE 5, e9851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mannella, P. & Brinton, R. D. Estrogen receptor protein interaction with phosphatidylinositol 3-kinase leads to activation of phosphorylated Akt and extracellular signal-regulated kinase 1/2 in the same population of cortical neurons: a unified mechanism of estrogen action. J. Neurosci. 26, 9439–9447 (2006). Demonstrates that oestradiol activates parallel neuroprotective signalling cascades in the same neuronal populations.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Jover-Mengual, T. et al. Acute estradiol protects CA1 neurons from ischemia-induced apoptotic cell death via the PI3K/Akt pathway. Brain Res. 1321, 1–12 (2010).

    Article  CAS  PubMed  Google Scholar 

  78. Garcia-Segura, L. M., Cardona-Gomez, P., Naftolin, F. & Chowen, J. A. Estradiol upregulates Bcl-2 expression in adult brain neurons. Neuroreport 9, 593–597 (1998).

    Article  CAS  PubMed  Google Scholar 

  79. Singer, C. A., Rogers, K. L. & Dorsa, D. M. Modulation of Bcl-2 expression: a potential component of estrogen protection in NT2 neurons. Neuroreport 9, 2565–2568 (1998).

    Article  CAS  PubMed  Google Scholar 

  80. Dubal, D. B., Shughrue, P. J., Wilson, M. E., Merchenthaler, I. & Wise, P. M. Estradiol modulates Bcl-2 in cerebral ischemia: a potential role for estrogen receptors. J. Neurosci. 19, 6385–6393 (1999).

    Article  CAS  PubMed  Google Scholar 

  81. Nilsen, J. & Diaz Brinton, R. Mechanism of estrogen-mediated neuroprotection: regulation of mitochondrial calcium and Bcl-2 expression. Proc. Natl Acad. Sci. USA 100, 2842–2847 (2003).

    Article  CAS  PubMed  Google Scholar 

  82. Jover-Mengual, T., Zukin, R. S. & Etgen, A. M. MAPK signaling is critical to estradiol protection of CA1 neurons in global ischemia. Endocrinology 148, 1131–1143 (2007).

    Article  CAS  PubMed  Google Scholar 

  83. Patrone, C., Andersson, S., Korhonen, L. & Lindholm, D. Estrogen receptor-dependent regulation of sensory neuron survival in developing dorsal root ganglion. Proc. Natl Acad. Sci. USA 96, 10905–10910 (1999).

    Article  CAS  PubMed  Google Scholar 

  84. Pike, C. J. Estrogen modulates neuronal Bcl-xL expression and β-amyloid-induced apoptosis: relevance to Alzheimer's disease. J. Neurochem. 72, 1552–1563 (1999).

    Article  CAS  PubMed  Google Scholar 

  85. Koski, C. L., Hila, S. & Hoffman, G. E. Regulation of cytokine-induced neuron death by ovarian hormones: involvement of antiapoptotic protein expression and c-JUN N-terminal kinase-mediated proapoptotic signaling. Endocrinology 145, 95–103 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Yao, M., Nguyen, T. V. & Pike, C. J. Estrogen regulates Bcl-w and Bim expression: role in protection against β-amyloid peptide-induced neuronal death. J. Neurosci. 27, 1422–1433 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Gollapudi, L. & Oblinger, M. M. Estrogen and NGF synergistically protect terminally differentiated, ERα-transfected PC12 cells from apoptosis. J. Neurosci. Res. 56, 471–481 (1999).

    Article  CAS  PubMed  Google Scholar 

  88. Zhao, L., Wu, T. W. & Brinton, R. D. Estrogen receptor subtypes α and β contribute to neuroprotection and increased Bcl-2 expression in primary hippocampal neurons. Brain Res. 1010, 22–34 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. Wu, T. W., Wang, J. M., Chen, S. & Brinton, R. D. 17β-estradiol induced Ca2+ influx via L-type calcium channels activates the Src/ERK/cyclic-AMP response element binding protein signal pathway and BCL-2 expression in rat hippocampal neurons: a potential initiation mechanism for estrogen-induced neuroprotection. Neuroscience 135, 59–72 (2005).

    Article  CAS  PubMed  Google Scholar 

  90. D'Astous, M., Mendez, P., Morissette, M., Garcia-Segura, L. M. & Di Paolo, T. Implication of the phosphatidylinositol-3 kinase/protein kinase B signaling pathway in the neuroprotective effect of estradiol in the striatum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice. Mol. Pharmacol. 69, 1492–1498 (2006).

    Article  CAS  PubMed  Google Scholar 

  91. Wang, S., Ren, P., Li, X., Guan, Y. & Zhang, Y. A. 17β-estradiol protects dopaminergic neurons in organotypic slice of mesencephalon by MAPK-mediated activation of anti-apoptosis gene BCL2. J. Mol. Neurosci. 45, 236–245 (2011).

    Article  CAS  PubMed  Google Scholar 

  92. Cardona-Rossinyol, A., Mir, M., Caraballo-Miralles, V., Llado, J. & Olmos, G. Neuroprotective effects of estradiol on motoneurons in a model of rat spinal cord embryonic explants. Cell. Mol. Neurobiol. 33, 421–432 (2013).

    Article  CAS  PubMed  Google Scholar 

  93. Dziennis, S., Jia, T., Ronnekleiv, O. K., Hurn, P. D. & Alkayed, N. J. Role of signal transducer and activator of transcription-3 in estradiol-mediated neuroprotection. J. Neurosci. 27, 7268–7274 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. De Butte-Smith, M., Zukin, R. S. & Etgen, A. M. Effects of global ischemia and estradiol pretreatment on phosphorylation of Akt, CREB and STAT3 in hippocampal CA1 of young and middle-aged female rats. Brain Res. 1471, 118–128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Sehara, Y. et al. Survivin is a transcriptional target of STAT3 critical to estradiol neuroprotection in global ischemia. J. Neurosci. 33, 12364–12374 (2013). Shows that ischaemia and oestradiol synergistically promote the activation of STAT3 and STAT3-dependent transcription of survivin . Inhibition or silencing of STAT3, or silencing of survivin , abolishes the neuroprotective action of oestradiol in the female rat hippocampus.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Seira, O. & Del Rio, J. A. Glycogen synthase kinase 3 β (GSK3β) at the tip of neuronal development and regeneration. Mol. Neurobiol. 49, 931–944 (2014).

    Article  CAS  PubMed  Google Scholar 

  97. Alonso, A. C., Zaidi, T., Grundke-Iqbal, I. & Iqbal, K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc. Natl Acad. Sci. USA 91, 5562–5566 (1994).

    Article  CAS  PubMed  Google Scholar 

  98. Cardona-Gomez, P., Perez, M., Avila, J., Garcia-Segura, L. M. & Wandosell, F. Estradiol inhibits GSK3 and regulates interaction of estrogen receptors, GSK3, and β-catenin in the hippocampus. Mol. Cell Neurosci. 25, 363–373 (2004).

    Article  CAS  PubMed  Google Scholar 

  99. Goodenough, S., Schleusner, D., Pietrzik, C., Skutella, T. & Behl, C. Glycogen synthase kinase 3β links neuroprotection by 17β-estradiol to key Alzheimer processes. Neuroscience 132, 581–589 (2005).

    Article  CAS  PubMed  Google Scholar 

  100. Perez-Alvarez, M. J., Maza Mdel, C., Anton, M., Ordonez, L. & Wandosell, F. Post-ischemic estradiol treatment reduced glial response and triggers distinct cortical and hippocampal signaling in a rat model of cerebral ischemia. J. Neuroinflamm. 9, 157 (2012).

    Article  CAS  Google Scholar 

  101. Varea, O. et al. Estradiol activates β-catenin dependent transcription in neurons. PLoS ONE 4, e5153 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Singh, M., Meyer, E. M. & Simpkins, J. W. The effect of ovariectomy and estradiol replacement on brain-derived neurotrophic factor messenger ribonucleic acid expression in cortical and hippocampal brain regions of female Sprague-Dawley rats. Endocrinology 136, 2320–2324 (1995).

    Article  CAS  PubMed  Google Scholar 

  103. Zhou, J., Zhang, H., Cohen, R. S. & Pandey, S. C. Effects of estrogen treatment on expression of brain-derived neurotrophic factor and cAMP response element-binding protein expression and phosphorylation in rat amygdaloid and hippocampal structures. Neuroendocrinology 81, 294–310 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Pietranera, L., Lima, A., Roig, P. & De Nicola, A. F. Involvement of brain-derived neurotrophic factor and neurogenesis in oestradiol neuroprotection of the hippocampus of hypertensive rats. J. Neuroendocrinol. 22, 1082–1092 (2010).

    Article  CAS  PubMed  Google Scholar 

  105. Luine, V. & Frankfurt, M. Interactions between estradiol, BDNF and dendritic spines in promoting memory. Neuroscience 239, 34–45 (2013).

    Article  CAS  PubMed  Google Scholar 

  106. Srivastava, D. P., Woolfrey, K. M. & Evans, P. D. Mechanisms underlying the interactions between rapid estrogenic and BDNF control of synaptic connectivity. Neuroscience 239, 17–33 (2013).

    Article  CAS  PubMed  Google Scholar 

  107. Harte-Hargrove, L. C., Maclusky, N. J. & Scharfman, H. E. Brain-derived neurotrophic factor-estrogen interactions in the hippocampal mossy fiber pathway: implications for normal brain function and disease. Neuroscience 239, 46–66 (2013).

    Article  CAS  PubMed  Google Scholar 

  108. Fernandez, A. M. & Torres-Aleman, I. The many faces of insulin-like peptide signalling in the brain. Nature Rev. Neurosci. 13, 225–239 (2012).

    Article  CAS  Google Scholar 

  109. Azcoitia, I., Sierra, A. & Garcia-Segura, L. M. Neuroprotective effects of estradiol in the adult rat hippocampus: interaction with insulin-like growth factor-I signalling. J. Neurosci. Res. 58, 815–822 (1999).

    Article  CAS  PubMed  Google Scholar 

  110. Perez-Martin, M., Azcoitia, I., Trejo, J. L., Sierra, A. & Garcia-Segura, L. M. An antagonist of estrogen receptors blocks the induction of adult neurogenesis by insulin-like growth factor-I in the dentate gyrus of adult female rat. Eur. J. Neurosci. 18, 923–930 (2003).

    Article  PubMed  Google Scholar 

  111. Quesada, A. & Micevych, P. E. Estrogen interacts with the IGF-1 system to protect nigrostriatal dopamine and maintain motoric behavior after 6-hydroxdopamine lesions. J. Neurosci. Res. 75, 107–116 (2004).

    Article  CAS  PubMed  Google Scholar 

  112. Selvamani, A. & Sohrabji, F. The neurotoxic effects of estrogen on ischemic stroke in older female rats is associated with age-dependent loss of insulin-like growth factor-1. J. Neurosci. 30, 6852–6861 (2010). Emphasizes the importance of oestradiol and IGF1 cross-talk in neuroprotection showing that oestradiol is protective against experimental stroke in young female mice but increases brain damage after stroke in aged mice due to decreased IGF1 levels.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Witty, C. F., Gardella, L. P., Perez, M. C. & Daniel, J. M. Short-term estradiol administration in aging ovariectomized rats provides lasting benefits for memory and the hippocampus: a role for insulin-like growth factor-I. Endocrinology 154, 842–852 (2013).

    Article  CAS  PubMed  Google Scholar 

  114. Nelson, B. S., Springer, R. C. & Daniel, J. M. Antagonism of brain insulin-like growth factor-1 receptors blocks estradiol effects on memory and levels of hippocampal synaptic proteins in ovariectomized rats. Psychopharmacol. 231, 899–907 (2014).

    Article  CAS  Google Scholar 

  115. Rettberg, J. R., Yao, J. & Brinton, R. D. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front. Neuroendocrinol. 35, 8–30 (2014).

    Article  CAS  PubMed  Google Scholar 

  116. Marin, R. et al. Role of estrogen receptor α in membrane-initiated signaling in neural cells: interaction with IGF-1 receptor. J. Steroid Biochem. Mol. Biol. 114, 2–7 (2009).

    Article  CAS  PubMed  Google Scholar 

  117. Mendez, P., Azcoitia, I. & Garcia-Segura, L. M. Estrogen receptor α forms estrogen-dependent multimolecular complexes with insulin-like growth factor receptor and phosphatidylinositol 3-kinase in the adult rat brain. Brain Res. Mol. Brain Res. 112, 170–176 (2003).

    Article  CAS  PubMed  Google Scholar 

  118. Toledo, E. M., Colombres, M. & Inestrosa, N. C. Wnt signaling in neuroprotection and stem cell differentiation. Prog. Neurobiol. 86, 281–296 (2008).

    Article  CAS  PubMed  Google Scholar 

  119. Lu, T. et al. REST and stress resistance in ageing and Alzheimer's disease. Nature 507, 448–454 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Zhang, Q. G., Wang, R., Khan, M., Mahesh, V. & Brann, D. W. Role of Dickkopf-1, an antagonist of the Wnt/β-catenin signaling pathway, in estrogen-induced neuroprotection and attenuation of tau phosphorylation. J. Neurosci. 28, 8430–8441 (2008). Details the regulation of WNT signalling by oestradiol: cerebral ischaemia activates JNK–JUN signalling, increasing the expression of the WNT inhibitor DKK1. Oestradiol reduces the activation of JNK–JUN signalling and downregulates the expression of DKK1. The decrease in DKK1 enhances neuroprotective WNT signalling, which in turn increases the expression of survivin , promoting neuronal survival.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Scott, E. L., Zhang, Q. G., Han, D., Desai, B. N. & Brann, D. W. Long-term estrogen deprivation leads to elevation of Dickkopf-1 and dysregulation of Wnt/β-Catenin signaling in hippocampal CA1 neurons. Steroids 78, 624–632 (2013).

    Article  CAS  PubMed  Google Scholar 

  122. Scali, C. et al. Inhibition of Wnt signaling, modulation of Tau phosphorylation and induction of neuronal cell death by DKK1. Neurobiol. Dis. 24, 254–265 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Wang, R. et al. Inhibition of MLK3-MKK4/7-JNK1/2 pathway by Akt1 in exogenous estrogen-induced neuroprotection against transient global cerebral ischemia by a non-genomic mechanism in male rats. J. Neurochem. 99, 1543–1554 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Ruiz-Palmero, I., Simon-Areces, J., Garcia-Segura, L. M. & Arevalo, M.-A. Notch/neurogenin 3 signalling is involved in the neuritogenic actions of oestradiol in developing hippocampal neurones. J. Neuroendocrinol. 23, 355–364 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Saldanha, C. J., Burstein, S. R. & Duncan, K. A. Induced synthesis of oestrogens by glia in the songbird brain. J. Neuroendocrinol. 25, 1032–1038 (2013).

    Article  CAS  PubMed  Google Scholar 

  126. Chamniansawat, S. & Chongthammakun, S. A priming role of local estrogen on exogenous estrogen-mediated synaptic plasticity and neuroprotection. Exp. Mol. Med. 44, 403–411 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Li, R. et al. Brain endogenous estrogen levels determine responses to estrogen replacement therapy via regulation of BACE1 and NEP in female Alzheimer's transgenic mice. Mol. Neurobiol. 47, 857–867 (2013).

    Article  CAS  PubMed  Google Scholar 

  128. Takahashi, K. et al. 11C-cetrozole: an improved C-11C-methylated PET probe for aromatase imaging in the brain. J. Nucl. Med. 55, 852–857 (2014).

    Article  CAS  PubMed  Google Scholar 

  129. Rodriguez-Navarro, J. A. et al. Gender differences and estrogen effects in parkin null mice. J. Neurochem. 106, 2143–2157 (2008).

    Article  CAS  PubMed  Google Scholar 

  130. Gillies, G. E. & McArthur, S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol. Rev. 62, 155–198 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Santos-Galindo, M., Acaz-Fonseca, E., Bellini, M. J. & Garcia-Segura, L. M. Sex differences in the inflammatory response of primary astrocytes to lipopolysaccharide. Biol. Sex. Differ. 2, 7 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Selvamani, A. & Sohrabji, F. Reproductive age modulates the impact of focal ischemia on the forebrain as well as the effects of estrogen treatment in female rats. Neurobiol. Aging 31, 1618–1628 (2010).

    Article  CAS  PubMed  Google Scholar 

  133. Zhang, Q. G. et al. C terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-α and the critical period hypothesis of estrogen neuroprotection. Proc. Natl Acad. Sci. USA 108, E617–624 (2011). Provides the molecular basis for the critical period hypothesis of oestradiol on neuroprotection, showing degradation of ERs in the hippocampus with ageing and after long-term oestradiol deprivation.

    Article  PubMed  Google Scholar 

  134. Diz-Chaves, Y. et al. Behavioral effects of estradiol therapy in ovariectomized rats depend on the age when the treatment is initiated. Exp. Gerontol. 47, 93–99 (2012).

    Article  CAS  PubMed  Google Scholar 

  135. Whitmer, R. A., Quesenberry, C. P., Zhou, J. & Yaffe, K. Timing of hormone therapy and dementia: the critical window theory revisited. Ann. Neurol. 69, 163–169 (2011).

    Article  PubMed  Google Scholar 

  136. Maki, P. M. Critical window hypothesis of hormone therapy and cognition: a scientific update on clinical studies. Menopause 20, 695–709 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Inagaki, T. & Etgen, A. M. Neuroprotective action of acute estrogens: animal models of brain ischemia and clinical implications. Steroids 78, 597–606 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Frank, A., Brown, L. M. & Clegg, D. J. The role of hypothalamic estrogen receptors in metabolic regulation. Front. Neuroendocrinol. 35, 550–557 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Tiwari-Woodruff, S., Morales, L. B., Lee, R. & Voskuhl, R. R. Differential neuroprotective and antiinflammatory effects of estrogen receptor (ER)α and ERβ ligand treatment. Proc. Natl Acad. Sci. USA 104, 14813–14818 (2007).

    Article  CAS  PubMed  Google Scholar 

  140. Nilsson, S., Koehler, K. F. & Gustafsson, J. A. Development of subtype-selective oestrogen receptor-based therapeutics. Nature Rev. Drug Discov. 10, 778–792 (2011).

    Article  CAS  Google Scholar 

  141. Chen, S., Ye, J., Kijima, I. & Evans, D. The HDAC inhibitor LBH589 (panobinostat) is an inhibitory modulator of aromatase gene expression. Proc. Natl Acad. Sci. USA 107, 11032–11037 (2010).

    Article  CAS  PubMed  Google Scholar 

  142. Yague, J. G., Garcia-Segura, L. M. & Azcoitia, I. Selective transcriptional regulation of aromatase gene by vitamin D, dexamethasone, and mifepristone in human glioma cells. Endocr. 35, 252–261 (2009).

    Article  CAS  Google Scholar 

  143. Midzak, A., Rone, M., Aghazadeh, Y., Culty, M. & Papadopoulos, V. Mitochondrial protein import and the genesis of steroidogenic mitochondria. Mol. Cell Endocrinol. 336, 70–79 (2011).

    Article  CAS  PubMed  Google Scholar 

  144. Melcangi, R. C., Garcia-Segura, L. M. & Mensah-Nyagan, A. G. Neuroactive steroids: state of the art and new perspectives. Cell. Mol. Life Sci. 65, 777–797 (2008).

    Article  CAS  PubMed  Google Scholar 

  145. Belelli, D. & Lambert, J. J. Neurosteroids: endogenous regulators of the GABAA receptor. Nature Rev. Neurosci. 6, 565–575 (2005).

    Article  CAS  Google Scholar 

  146. Schule, C., Nothdurfter, C. & Rupprecht, R. The role of allopregnanolone in depression and anxiety. Prog. Neurobiol. 113, 79–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  147. Azcoitia, I., Yague, J. G. & Garcia-Segura, L. M. Estradiol synthesis within the human brain. Neuroscience 191, 139–147 (2011).

    Article  CAS  PubMed  Google Scholar 

  148. Schlinger, B. A. & Callard, G. V. Localization of aromatase in synaptosomal and microsomal subfractions of quail (Coturnix coturnix japonica) brain. Neuroendocrinology 49, 434–441 (1989).

    Article  CAS  PubMed  Google Scholar 

  149. Naftolin, F. et al. Aromatase immunoreactivity in axon terminals of the vertebrate brain. An immunocytochemical study on quail, rat, monkey and human tissues. Neuroendocrinology 63, 149–155 (1996).

    Article  CAS  PubMed  Google Scholar 

  150. Hojo, Y. et al. Adult male rat hippocampus synthesizes estradiol from pregnenolone by cytochromes P45017α and P450 aromatase localized in neurons. Proc. Natl Acad. Sci. USA 101, 865–870 (2004).

    Article  CAS  PubMed  Google Scholar 

  151. Vierk, R. et al. Aromatase inhibition abolishes LTP generation in female but not in male mice. J. Neurosci. 32, 8116–8126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Bian, C., Zhu, H., Zhao, Y., Cai, W. & Zhang, J. Intriguing roles of hippocampus-synthesized 17β-estradiol in the modulation of hippocampal synaptic plasticity. J. Mol. Neurosci. 54, 271–281 (2014).

    Article  CAS  PubMed  Google Scholar 

  153. Fester, L. et al. Proliferation and apoptosis of hippocampal granule cells require local oestrogen synthesis. J. Neurochem. 97, 1136–1144 (2006).

    Article  CAS  PubMed  Google Scholar 

  154. Bowers, J. M., Waddell, J. & McCarthy, M. M. A developmental sex difference in hippocampal neurogenesis is mediated by endogenous oestradiol. Biol. Sex. Differ. 1, 8 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Cornil, C. A., Ball, G. F. & Balthazart, J. Rapid control of male typical behaviors by brain-derived estrogens. Front. Neuroendocrinol. 33, 425–446 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Liu, N. J., Chakrabarti, S., Schnell, S., Wessendorf, M. & Gintzler, A. R. Spinal synthesis of estrogen and concomitant signaling by membrane estrogen receptors regulate spinal κ- and μ-opioid receptor heterodimerization and female-specific spinal morphine antinociception. J. Neurosci. 31, 11836–11845 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Ghorbanpoor, S., Garcia-Segura, L. M., Haeri-Rohani, A., Khodagholi, F. & Jorjani, M. Aromatase inhibition exacerbates pain and reactive gliosis in the dorsal horn of the spinal cord of female rats caused by spinothalamic tract injury. Endocrinology 155, 4341–4355 (2014).

    Article  CAS  PubMed  Google Scholar 

  158. Remage-Healey, L., Dong, S. M., Chao, A. & Schlinger, B. A. Sex-specific, rapid neuroestrogen fluctuations and neurophysiological actions in the songbird auditory forebrain. J. Neurophysiol. 107, 1621–1631 (2012).

    Article  CAS  PubMed  Google Scholar 

  159. Dalla, C., Antoniou, K., Papadopoulou-Daifoti, Z., Balthazart, J. & Bakker, J. Oestrogen-deficient female aromatase knockout (ArKO) mice exhibit depressive-like symptomatology. Eur. J. Neurosci. 20, 217–228 (2004).

    Article  CAS  PubMed  Google Scholar 

  160. Alejandre-Gomez, M., Garcia-Segura, L. M. & Gonzalez-Burgos, I. Administration of an inhibitor of estrogen biosynthesis facilitates working memory acquisition in male rats. Neurosci. Res. 58, 272–277 (2007).

    Article  CAS  PubMed  Google Scholar 

  161. Bailey, D. J., Ma, C., Soma, K. K. & Saldanha, C. J. Inhibition of hippocampal aromatization impairs spatial memory performance in a male songbird. Endocrinology 154, 4707–4714 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Prange-Kiel, J. et al. Gonadotropin-releasing hormone regulates spine density via its regulatory role in hippocampal estrogen synthesis. J. Cell Biol. 180, 417–426 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Remage-Healey, L., Saldanha, C. J. & Schlinger, B. A. Estradiol synthesis and action at the synapse: evidence for “synaptocrine” signaling. Front. Endocrinol. 2, 28 (2011).

    Article  Google Scholar 

  164. Maximov, P. Y., Lee, T. M. & Jordan, V. C. The discovery and development of selective estrogen receptor modulators (SERMs) for clinical practice. Curr. Clin. Pharmacol. 8, 135–155 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Bourque, M., Morissette, M. & Di Paolo, T. Raloxifene activates G protein-coupled estrogen receptor 1/Akt signaling to protect dopamine neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice. Neurobiol. Aging 35, 2347–2356 (2014).

    Article  CAS  PubMed  Google Scholar 

  166. Velazquez-Zamora, D. A., Garcia-Segura, L. M. & Gonzalez-Burgos, I. Effects of selective estrogen receptor modulators on allocentric working memory performance and on dendritic spines in medial prefrontal cortex pyramidal neurons of ovariectomized rats. Horm. Behav. 61, 512–517 (2012).

    Article  CAS  PubMed  Google Scholar 

  167. Walf, A. A. & Frye, C. A. Raloxifene and/or estradiol decrease anxiety-like and depressive-like behavior, whereas only estradiol increases carcinogen-induced tumorigenesis and uterine proliferation among ovariectomized rats. Behav. Pharmacol. 21, 231–240 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Calmarza-Font, I., Lagunas, N. & Garcia-Segura, L. M. Antidepressive and anxiolytic activity of selective estrogen receptor modulators in ovariectomized mice submitted to chronic unpredictable stress. Behav. Brain Res. 227, 287–290 (2012).

    Article  CAS  PubMed  Google Scholar 

  169. Barreto, G. et al. Selective estrogen receptor modulators decrease reactive astrogliosis in the injured brain: effects of aging and prolonged depletion of ovarian hormones. Endocrinology 150, 5010–5015 (2009).

    Article  CAS  PubMed  Google Scholar 

  170. Barreto, G. E., Santos-Galindo, M. & Garcia-Segura, L. M. Selective estrogen receptor modulators regulate reactive microglia after penetrating brain injury. Front. Aging Neurosci. 6, 132 (2014).

    PubMed  PubMed Central  Google Scholar 

  171. Mosquera, L. et al. Tamoxifen and estradiol improved locomotor function and increased spared tissue in rats after spinal cord injury: their antioxidant effect and role of estrogen receptor α. Brain Res. 1561, 11–22 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Sun, X., Ji, C., Hu, T., Wang, Z. & Chen, G. Tamoxifen as an effective neuroprotectant against early brain injury and learning deficits induced by subarachnoid hemorrhage: possible involvement of inflammatory signaling. J. Neuroinflamm. 10, 157 (2013).

    Article  CAS  Google Scholar 

  173. Khan, M. M., Wakade, C., de Sevilla, L. & Brann, D. W. Selective estrogen receptor modulators (SERMs) enhance neurogenesis and spine density following focal cerebral ischemia. J. Steroid Biochem. Mol. Biol. http://dx.doi.org/10.1016/j.jsbmb.2014.05.001 (2014).

  174. Rzemieniec, J. et al. Neuroprotective action of raloxifene against hypoxia-induced damage in mouse hippocampal cells depends on ERα but not ERβ or GPR30 signalling. J. Steroid Biochem. Mol. Biol. http://dx.doi.org/10.1016/j.jsbmb.2014.05.005 (2014).

  175. Castello-Ruiz, M. et al. The selective estrogen receptor modulator, bazedoxifene, reduces ischemic brain damage in male rat. Neurosci. Lett. 575, 53–57 (2014).

    Article  CAS  PubMed  Google Scholar 

  176. Tapia-Gonzalez, S., Carrero, P., Pernia, O., Garcia-Segura, L. M. & Diz-Chaves, Y. Selective oestrogen receptor (ER) modulators reduce microglia reactivity in vivo after peripheral inflammation: potential role of microglial ERs. J. Endocrinol. 198, 219–230 (2008).

    Article  CAS  PubMed  Google Scholar 

  177. Bebo, B. F. Jr et al. Treatment with selective estrogen receptor modulators regulates myelin specific T-cells and suppresses experimental autoimmune encephalomyelitis. Glia 57, 777–790 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  178. Li, R. et al. Raloxifene suppresses experimental autoimmune encephalomyelitis and NF-κB-dependent CCL20 expression in reactive astrocytes. PLoS ONE 9, e94320 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Carroll, J. C. & Pike, C. J. Selective estrogen receptor modulators differentially regulate Alzheimer-like changes in female 3xTg-AD mice. Endocrinology 149, 2607–2611 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Herrera, J. L., Fernandez, C., Diaz, M., Cury, D. & Marin, R. Estradiol and tamoxifen differentially regulate a plasmalemmal voltage-dependent anion channel involved in amyloid-β induced neurotoxicity. Steroids 76, 840–844 (2011).

    CAS  PubMed  Google Scholar 

  181. Bourque, M., Liu, B., Dluzen, D. E. & Di Paolo, T. Tamoxifen protects male mice nigrostriatal dopamine against methamphetamine-induced toxicity. Biochem. Pharmacol. 74, 1413–1423 (2007).

    Article  CAS  PubMed  Google Scholar 

  182. Arevalo, M.-A., Santos-Galindo, M., Lagunas, N., Azcoitia, I. & Garcia-Segura, L. M. Selective estrogen receptor modulators as brain therapeutic agents. J. Mol. Endocrinol. 46, R1–R9 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank M. Garcia-Diaz, Stony Brook University School of Medicine, New York, USA, for critical reading of the manuscript. The authors acknowledge support from Ministerio de Economía y Competitividad, Spain (BFU2011-30217-C03-01).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Luis M. Garcia-Segura.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Hormone

A cellular secretion released into circulation that targets multiple cell types in multiple organs.

Autocrine factor

A substance released by a cell that targets nearby cells of the same type.

Paracrine factor

A substance released by a cell that targets nearby cells of multiple types.

Excitotoxic injury

A pathological process by which neurons are damaged and killed by the overactivation of glutamate receptors.

Antisense oligonucleotides

Oligonucleotides with a sequence that is complementary to the mRNA of a given molecule, which can be used to block its translation. The subsequent temporary elimination of the protein of interest often provides useful information on its biological function.

Transcriptional co-activators and co-repressors

Proteins that activate or repress transcriptional activity by binding to transcription factors. Co-activators and co-repressors are unable to bind to DNA by themselves.

Lipid rafts

Submicrodomains of the plasma membrane that are enriched in sphingolipids and cholesterol and are thought to serve as a signalling platform.

Phosphoinositide 3-kinase (PI3K) signalling pathway

A cell survival pathway that is activated by receptor tyrosine kinases. The activation of PI3K results in the phosphorylation and activation of the kinase AKT and in the subsequent phosphorylation and inhibition of glycogen synthase kinase-3β.

Neurovascular unit

A complex multicellular functional unit of the CNS comprising vascular cells, glial cells and neurons, which together determine CNS activities and responses in health and disease.

Coincidence signal detector

A mechanism that detects the temporal coincidence of multiple signals.

Positron emission tomography

A medical imaging technique that uses injected radiolabelled tracer compounds, in conjunction with mathematical reconstruction methods, to produce a three-dimensional image, or map, of functional processes in the body (such as glucose metabolism, blood flow or receptor distributions).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arevalo, MA., Azcoitia, I. & Garcia-Segura, L. The neuroprotective actions of oestradiol and oestrogen receptors. Nat Rev Neurosci 16, 17–29 (2015). https://doi.org/10.1038/nrn3856

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn3856

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing