Viewpoint
The female sex hormone oestrogen as a neuroprotectant

https://doi.org/10.1016/S0165-6147(99)01392-9Get rights and content

Abstract

It is well recognized that oestrogen regulates sex differentiation and maturation of sex organs via binding to specific intracellular receptors. However, oestrogen receptors (ERs) are expressed in a variety of other tissues, including the nervous system, which suggests that oestrogen’s effects are not limited to primary and secondary sex organs. Increasing evidence supports the role of oestrogen as a neuroprotective compound that can act dependently or independently of ER activation; oestrogen has recently been shown to exhibit intrinsic antioxidant activity that is ER independent. Thus, oestrogen might represent a potential ‘chemical shield’ for neurones. In this article, some recent advances in the elucidation of oestrogen’s beneficial activities on nerve cell survival are discussed.

Section snippets

Direct modulation of neuronal gene transcription by oestrogens

In the mammalian brain, activation of oestrogen receptors (ERs) has long-term genomic effects such as regulation of nerve cell development and protection of the brain during aging2, which helps to preserve memory and other cognitive functions that are assigned mainly to the hippocampus. Interestingly, the actions of oestrogens lead to sex differences in the severity of brain damage resulting from transient ischaemia, and in the response of the brain to lesions and chronic stress2. The genomic

Interaction of oestrogen with neuronal membranes

Not all of the neuromodulatory activities of oestrogens are a result of nuclear receptor occupation, receptor activation and alteration of gene transcription2. For example, in addition to the classical ERs, possible membrane-binding sites (membrane receptors) for various steroids, including oestrogens, are being studied intensively16. Recently, in vitro data indicate that both membrane and nuclear ERs can be derived from a single transcript13. Indeed, oestrogens can also affect neurones via

Oestrogens and neurodegenerative disorders

Depletion of oestrogen in the female rat has been associated with changes in neuronal plasticity and, in general, oestrogens appear to have beneficial effects on mental performance2. The decrease in oestrogen levels after the menopause is associated with increased frequency of AD; replacement with oestrogen reduces the risk of women developing AD (28, 29). As a consequence of the oestrogen decline after the menopause, the age-related increased incidence in neurodegenerative disorders and the

Concluding remarks

The steroid hormone oestrogen is now recognized to possess many actions. Oestrogen is sexually dimorphic; it is not restricted to the female as the male sex hormone testosterone [and other steroids that contain a 19-carbon atom structure (C-19 steroids)] can be converted locally to oestradiol in various tissues, including the brain, by an aromatase cytochrome P450 enzyme. The ovaries remain the main source of oestrogen; however, the existence of multiple sites of activity, including the brain,

References (39)

  • L.M. Mudd et al.

    Brain Res. Bull.

    (1998)
  • M.J. Kelly et al.

    Steroids

    (1999)
  • V.D. Ramirez et al.

    Front. Neuroendocrinol.

    (1996)
  • H.H. Zakon

    Trends Neurosci.

    (1998)
  • R. Rupprecht et al.

    Trends Neurosci.

    (1999)
  • C. Behl et al.

    Biochem. Biophys. Res. Commun.

    (1995)
  • M.X. Tang

    Lancet

    (1996)
  • J. Hardy

    Trends Neurosci.

    (1997)
  • C. Behl

    Progr. Neurobiol.

    (1999)
  • N.L. Weigel

    Biochem. J.

    (1996)
  • B.S. McEwen et al.

    Endocr. Rev.

    (1999)
  • C.D. Toran-Allerand

    Dev. Neurosci.

    (1996)
  • P.J. Shugrue et al.

    J. Comp. Neurol.

    (1997)
  • S. Nilsson et al.

    Trends Endocrinol. Metab.

    (1997)
  • J.J. Watters et al.

    J. Neurosci.

    (1998)
  • B. Stein et al.

    Mol. Cell. Biol.

    (1995)
  • R. Galien et al.

    Nucleic Acids Res.

    (1997)
  • T. Improta-Brears

    Proc. Natl. Acad. Sci. U. S. A.

    (1999)
  • C.A. Singer et al.

    J. Neurosci.

    (1999)
  • Cited by (123)

    • Neuroprotection with Estradiol in Experimental Perinatal Asphyxia: A New Approach. A New Approach.

      2015, Estrogen Effects on Traumatic Brain Injury: Mechanisms of Neuroprotection and Repair
    • Prometaphase arrest-dependent phosphorylation of Bcl-2 family proteins and activation of mitochondrial apoptotic pathway are associated with 17α-estradiol-induced apoptosis in human Jurkat T cells

      2013, Biochimica et Biophysica Acta - Molecular Cell Research
      Citation Excerpt :

      The biological activity of estrogens is mediated by three different mechanisms: the intracellular estrogen receptor (ER)-mediated genomic mechanism; the plasma membrane ER-mediated nongenomic mechanism associated with cell signaling pathways; and an ER-independent mechanism [1,2].

    • Role of protein phosphatases and mitochondria in the neuroprotective effects of estrogens

      2009, Frontiers in Neuroendocrinology
      Citation Excerpt :

      The antioxidant activity of estrogens observed at high (non-physiological) concentrations (10−5 M) is dependent upon the presence of a phenolic A ring in the steroid structure and is independent of activation of the ERs [10–12,36,110]. A large body of evidence has demonstrated that estradiol possesses antioxidant properties and suppresses oxidative stress in neurons and neuronal cell lines induced by hydrogen peroxide, superoxide anions and other pro-oxidants [8–12,18,23,151]. Both 17α- and 17β-estradiol have similar antioxidant effects [11,12,18,61], suggesting that the neuroprotective effects of estrogens are in part ER-independent.

    • Estradiol and neurodegenerative oxidative stress

      2008, Frontiers in Neuroendocrinology
      Citation Excerpt :

      17β-Estradiol (referred to as estradiol in the rest of the text) treatment can protect against a wide range of toxic insults including free radical generators [12,21,60] excitotoxicity [21,136,138], β amyloid-induced toxicity [21] and ischemia [45,59].

    View all citing articles on Scopus
    View full text