Elsevier

Brain Research Reviews

Volume 57, Issue 2, 14 March 2008, Pages 431-443
Brain Research Reviews

Review
Loss of aromatase cytochrome P450 function as a risk factor for Parkinson's disease?

https://doi.org/10.1016/j.brainresrev.2007.10.011Get rights and content

Abstract

The final step in the physiological synthesis of 17β estradiol (E2) is aromatization of precursor testosterone by a CYP19 gene product, cytochrome P450 estrogen aromatase in the C19 steroid metabolic pathway. Within the central nervous system (CNS) the presence, distribution, and activity of aromatase have been well characterized. Developmental stage and injury are known modulators of brain enzyme activity, where both neurons and glial cells reportedly have the capability to synthesize this key estrogenic enzyme. The gonadal steroid E2 is a critical survival, neurotrophic and neuroprotective factor for dopaminergic neurons of the substantia nigra pars compacta (SNpc), the cells that degenerate in Parkinson's disease (PD). In previous studies we underlined a crucial role for the estrogenic status at the time of injury in dictating vulnerability to the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our ongoing studies address the contribution of brain aromatase and extragonadal E2 as vulnerability factors for PD pathology in female brain, by exposing aromatase knockout (ArKO, −/−) female mice which are unable to synthesize estrogens to MPTP. Our initial results indicate that aromatase deficiency from early embryonic life significantly impairs the functional integrity of SNpc tyrosine hydroxylase-positive neurons and dopamine transporter innervation of the caudate–putamen in adulthood. In addition, ArKO females exhibited a far greater vulnerability to MPTP-induced nigrostriatal damage as compared to their Wt type gonadally intact and gonadectomized counterparts. Characterization of this novel implication of P450 aromatase as determining factor for PD vulnerability may unravel new avenues for the understanding and development of novel therapeutic approaches for Parkinson's disease.

Introduction

The female hormone, 17β estradiol (E2), plays a fundamental role in a multitude of physiological processes in mammals, including reproduction, cardiovascular health, skeletal growth and bone homeostasis, immune and cognitive functions. Within the brain, E2 plays a pivotal role in the regulation of neuron survival, differentiation and plasticity (McEwen and Alves, 1999, Garcia-Segura et al., 2001, Morris et al., 2004). One key aspect is represented by the critical action of E2 in dictating gender-specific processes of brain development (Wilson and Davies, 2007). Indeed, the conversion of androgens to estrogens within the CNS is a critical means by which testosterone regulates many physiological and behavioural processes such as sexual differentiation of the brain (MacLusky and Naftolin, 1981). This critical step is performed by aromatase, a member of the P450 superfamily of cytochrome enzymes, and a product of the CYP19 gene (Roselli and Klosterman, 1998, Bakker et al., 2003). Unlike other P450 enzyme members, aromatase is the only one capable of creating an aromatic ring characterizing the estrogenic structure.

Aromatase is tissue-specifically regulated in various organs and cells and plays an important role through endocrine and intracrine estrogen synthesis (Harada et al., 1993, Honda et al., 1994, Simpson and Davis, 2001). Therefore aromatase deficiency causes crucial impairments of physiological functions in both gonadal and extragonadal tissues. In the CNS, aromatase has been shown to regulate neural plasticity by stimulating growth and migration of cells, protecting against degeneration and brain injury, regulating the reproductive endocrine axis, influencing learning and memory processes, stress and mood (Lephart et al., 2001). Of special interest, remarkable neuroprotective effects of brain aromatase have been demonstrated with respect to kainic acid excitotoxicity (Garcia-Segura et al., 1999), stroke (McCullough et al., 2003), Alzheimer's (ALZ) disease and epilepsy (Yue et al., 2005). In addition, polymorphisms in the CYP19 gene were shown to confer increased risk for ALZ pathology (Ivonen et al., 2004, Huang and Poduslo, 2006). While in normal conditions brain aromatase is almost exclusively localized in neurons, brain injury up-regulates this estrogenic enzymatic activity in astrocytes, and the implication of increased glial aromatase for neuroprotection has been highlighted in the studies of Garcia-Segura and collaborators (Garcia-Segura et al., 1999, Azcoitia et al., 2001, Garcia-Segura et al., 2001, Garcia-Ovejero et al., 2005).

In the brain, the abundance and activity of aromatase in rodents and humans have been well characterized (Lephart et al., 2001). In particular, aromatase-positive neurons have been localized within the lateral septal region, the bed nucleus of the stria terminalis, the hippocampus, the amygdala, several hypothalamic nuclei, the medial preoptic area, the nucleus accumbens, and several regions of the cortex, such as in the piriform lobe (Balthazart et al., 1991, Balthazart and Ball, 1998, Foidart and Harada, 1995, Jakab et al., 1994).

Aromatase has been also documented to participate in the physiological development of midbrain dopaminergic neurons in embryonic and early neonatal brain (Kipp et al., 2006). Indeed, E2 is a specific physiological regulator of nigrostriatal dopaminergic neurons during development, in adulthood as well as during neuronal degenerative processes, such as Parkinson's disease (PD) (Beyer and Karolezak, 2000, Dluzen, 2000, Leranth et al., 2000, Grandbois et al., 2000, Kuppers et al., 2000, Dluzen and Horstink, 2003, D'Astous et al., 2004a, D'Astous et al., 2004b, Kipp et al., 2006, Morale et al., 2006), a progressive degenerative disorder characterized by the selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) (Olanow et al., 2003).

In the present work, after a brief review of major risk and neuroprotective factors controlling vulnerability to PD, the evidence linking brain E2 synthesis within the ventral midbrain and striatum will be reviewed and the hypothesis of brain aromatase deficiency as a risk factor for the vulnerability to PD pathology presented using a genetic approach, i.e. the aromatase knockout mouse strain, generated by Honda et al. (1998). Aromatase activity is sexually differentiated in mice during neonatal period as well as in adulthood (Bakker et al., 2004a, Bakker et al., 2004b) with higher testosterone-stimulated activity in males than in females. However, the neonatal female brain produces substantial amounts of estrogen that could play a significant role not only during the sexual differentiation of the female brain early in life (see Bakker et al., 2004a, Bakker et al., 2004b) but also later in life in modulating the future ability of brain cells to respond to brain injury (McCullough et al., 2003, Yue et al., 2005). Absence of aromatase activity in the brain and gonads of these ArKO mice (Bakker et al., 2004a, Bakker et al., 2004b) validates the ArKO mouse as a valuable tool in the study of the role of estradiol in modulating the vulnerability of nigrostriatal dopaminergic neurons to the environmental neurotoxin, MPTP.

Section snippets

Complex interplay between genetic and environmental factors modulate vulnerability to Parkinson's disease

Mesencephalic dopaminergic neurons are among the elements of the basal ganglia most vulnerable to neurodegeneration. Within degenerative CNS disorders, PD is the second most frequent pathology after Alzheimer's disease. The loss of dopaminergic afferents to the striatum and putamen results in extrapyramidal motor dysfunction, including tremor, rigidity, and bradykinesia accompanied by progressive impairment of autonomic, cognitive and mood functions (Di Monte and Langston, 1995, Olanow et al.,

Aromatase cytochrome P450 knock out (ArKO) mice as models to study the impact of lifelong brain estrogen deficiency

Mice with targeted disruption of exon 9 of the CYP19 gene [aromatase knockout (ArKO)] have been developed by several laboratories (Fisher et al., 1998, Honda et al., 1998, Toda et al., 2001). Unlike other models of E2 insufficiency (e.g. ER knockout mice), ArKOs cannot synthesize E2 but retain the ability to respond to exogenous estrogen because of the expression of ERs. The ArKO phenotype is characterized by low plasma estradiol, elevated testosterone, small atrophic uteri, decreased

Modeling the role of aromatase deficiency in Parkinson's disease

The MPTP-induced model of striatal dopamine depletion has been extensively used to elucidate the determinants of PD pathology in rodents and nonhuman primates. There are some important aspects to underline in order to develop successful MPTP mouse models of PD that have been recently reviewed by Jasckson-Lewis and Przedborski, (2007). It should be anticipated that the most relevant regimens of MPTP are those that create an overt and stable lesion of the nigrostriatal pathway with the least

Lifelong aromatase deficiency impairs nigrostriatal dopaminergic neurons integrity and exacerbates vulnerability to MPTP

To investigate potential changes in the density of TH+ SNpc neurons, we counted, on serial sections, the total number of TH+ cells throughout the entire rostro-caudal (RC) axis of the murine SNpc (Franklin and Paxinos, 1997). Wt and ArKO mice were sacrificed 7 days after the last saline or MPTP injections, as previously described (Morale et al. 2004). Within the SNpc, TH-immunofluorescent (IF) profiles (Fig. 1B), as well as TH- and DAT-IF nigrostriatal projections (Fig. 2C–D) differed between

Conclusion

Altogether the information herein presented support a pivotal role for brain aromatase activity not only during ontogenic development of the nigrostriatal dopaminergic system, but also in adult life, when brain E2 synthesis might contribute to the protection of nigral dopaminergic neurons against neurotoxic insults. During menopause, long-term deprivation of circulating E2 and decrease of brain aromatase activity may expose the aging brain to several insults, including increased risk/incidence

Acknowledgments

The work was supported by grants of the Italian Ministry of Health (RF-2005-07 - Agreement no. 05.82) and Italian Ministry of Research. Authors are grateful to Dr. J. Bakker, University of Liege (Belgium) for her help in establishing our mice colony.

References (122)

  • HaradaN. et al.

    Analysis of spatiotemporal regulation of aromatase in the brain using transgenic mice

    Steroid Biochem. Mol. Biol.

    (2005)
  • HillR.A. et al.

    Estrogen deficiency leads to apoptosis in dopaminergic neurons in the medial preoptic area and arcuate nucleus of male mice

    Mol. Cell Neurosci.

    (2004)
  • HondaS. et al.

    Disruption of sexual behavior in male aromatase-deficient mice lacking exons 1 and 2 of the Cyp19 gene

    Biochem. Biophys. Res. Commun.

    (1998)
  • HondaS. et al.

    Novel exon 1 of the aromatase gene specific for aromatase transcripts in human brain

    Biochem. Biophys. Res. Commun.

    (1994)
  • JakabR.L. et al.

    Aromatase (estrogen synthase) immunoreactive neurons in the rat septal area. A light and electron microscopic study

    Brain Res.

    (1994)
  • KippM. et al.

    Estrogen and the development and protection of nigrostriatal dopaminergic neurons: concerted action of a multitude of signals, protective molecules, and growth factors

    Front. Neuroendocrinol.

    (2006)
  • KuppersE. et al.

    Expression of aromatase in the embryonic and post-natal mouse striatum

    Mol. Brain Res.

    (1998)
  • LephartE.D. et al.

    Brain androgen and progesterone metabolizing enzymes : biosynthesis, distribution and function

    Brain Res. Rev.

    (2001)
  • MacLuskyN.J. et al.

    Aromatase in the cerebral cortex, hippocampus and midbrain: ontogeny and developmental implications

    Mol. Cell. Neurosci.

    (1994)
  • MarchettiB. et al.

    To be or not to be (inflamed) — is that the question in anti-inflammatory drug therapy of neurodegenerative disorders?

    Trends Pharmacol. Sci.

    (2005)
  • MarchettiB. et al.

    Stress, the immune system and vulnerability to degenerative disorders of the central nervous system in transgenic mice expressing glucocorticoid receptor antisense RNA

    Brain Res. Rev.

    (2001)
  • MarchettiB. et al.

    Glucocorticoid receptor-nitric oxide crosstalk and vulnerability to experimental Parkinsonism: pivotal role for glia-neuron interactions

    Brain Res. Rev.

    (2005)
  • McGeerP.L. et al.

    Inflammation and neurodegeneration in Parkinson's disease

    Parkinsonism Relat. Disord.

    (2004)
  • MoraleM.C. et al.

    Estrogen, neuroinflammation and neuroprotection in Parkinson's disease: glia dictates resistance versus vulnerability to neurodegeneration

    Neuroscience

    (2006)
  • RaabH. et al.

    Ontogeny of aromatase messenger ribonucleic acid and aromatase activity in the rat midbrain

    Mol. Brain Res

    (1995)
  • RaabH. et al.

    Ontogenic expression and splicing of estrogen receptor-alpha and beta mRNA in the rat brain

    Neurosci. Lett.

    (1999)
  • AscherioA. et al.

    Caffeine, post-menopausal estrogen and risk of Parkinson's disease

    Neurology

    (2003)
  • BakkerJ. et al.

    The aromatase knockout mouse provides new evidence that estradiol is required during development in the female for the expression of socio-sexual behaviours in adulthood

    J. Neurosci.

    (2002)
  • BakkerJ. et al.

    The aromatase knockout (ArKO) mouse provides new evidence that estrogens are required for the development of the female brain

    Ann. N. Y. Acad. Sci.

    (2003)
  • BakkerJ. et al.

    Relationships between aromatase activity in the brain and gonads and behavioural deficits in homozygous and heterozygous aromatase knockout mice

    J. Neuroendocrinol.

    (2004)
  • BalthazartJ. et al.

    Distribution of aromatase-immunoreactive cells in the mouse forebrain

    Cell Tissue Res.

    (1991)
  • BenedettiM.D. et al.

    Histerectomy, menopause, and estrogen use preceding Parkinson's disease: an exploratory case study

    Mov. Disord.

    (2001)
  • BetarbetR. et al.

    Chronic systemic pesticide exposure reproduces features of Parkinson's disease

    Nat. Neurosci.

    (2000)
  • BeyerC.

    Estrogen and the developing mammalian brain

    Anat. Embryol.

    (1999)
  • BeyerC. et al.

    Estrogenic stimulation of neurite outgrowth in midbrain dopaminergic neurons depends on cAMP/protein kinase A signalling

    J. Neurosci. Res.

    (2000)
  • BeyerC. et al.

    Regulation of gene expression in the developing midbrain by estrogen: implication of classical and nonclassical steroid signaling

    Ann. NY. Acad. Sci.

    (2003)
  • BezardE.S. et al.

    Enriched environment confers resistance to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and cocaine: involvement of dopamine transporter and trophic factors

    J. Neurosci.

    (2003)
  • BlanchetP.J. et al.

    Short-term effects of high dose17-beta-estradiol in post-menopausal PD patients: a crossover study

    Neurology

    (1999)
  • BrintonR.D.

    Cellular and molecular mechanisms of estrogen regulation of memory function and neuroprotection against Alzheimer's disease: recent insight and remaining challenges

    Learn. Mem.

    (2001)
  • BrintonR.D.

    Requirements of a brain selective estrogen: advances and remaining challenges for developing a NeuroSERM

    J. Alzheimers Dis.

    (2004)
  • BurnsR.S. et al.

    A primate model of Parkinsonism: selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydroperidine

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

    (1983)
  • CallierS. et al.

    Stereospecific prevention by 17-beta estradiol of MPTP-induced dopamine depletion

    Synapse

    (2000)
  • Cantutti-CastelvetriI. et al.

    Effect of gender on nigral gene expression and Parkinson's disease

    Neurobiol. Dis.

    (2007)
  • L.M. Creutz et al.

    Estrogen receptor-beta immunoreactivity in the midbrain of adult rats: regional, subregional, and cellular localization in the A10, A9, and A8 dopamine cell groups

    J. Comp. Neurol.

    (2002)
  • CreutzL.M. et al.

    Mesostriatal and mesolimbic projections of midbrain neurons immunoreactive for estrogen receptor beta or androgen receptors in rats

    J. Comp. Neurol.

    (2004)
  • CurrieL.J. et al.

    Postmenopausal estrogen use affects risk for Parkinson's disease

    Neurology

    (2004)
  • DallaC. et al.

    Oestrogen-deficient female aromatase-knockout (ArKO) mice exhibit depressive-like symptomatology

    Eur. J. Neurosci.

    (2004)
  • D'AstousM. et al.

    Dopamine transporter as marker of neuroprotection in methamphetamine-lesioned mice treated acutely with estradiol

    Neuroendocrinology

    (2004)
  • DiamondS.G. et al.

    An examination of male–female differences in progression and mortality of Parkinson's disease

    Neurology

    (1990)
  • Di MonteD.A. et al.

    Idiopathic and 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism

  • Cited by (57)

    • Effective anti-aromatase therapy to battle against estrogen-mediated breast cancer: Comparative SAR/QSAR assessment on steroidal aromatase inhibitors

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Therefore, apart from the regulation of fertility and reproduction, it is found to modulate the cell growth, migration, neuroplasticity, neuroprotection, and brain injury [39]. It has also been found to take part in epileptic conditions, Parkinson’s and Alzheimer’s disease [40–42]. During the biosynthesis of estrogen, aromatase enzyme plays a major role to develop breast cancer through a rate limiting kinetics [7].

    • Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease

      2020, Redox Biology
      Citation Excerpt :

      Interestingly, looking at gene expression profiling in SN of female and male post mortem PD brain, Cantuti-Castelvetri [212] documented that genes upregulated in females relative to males are mainly involved in signal transduction and neuronal maturation, protein kinase activity, and Wnt signaling pathway [212]. Importantly, sex steroids, particularly estrogens, are important in protecting midbrain astrocytes and dopaminergic neurons against oxidative and inflammatory insult, whereas their abrogation in estrogen-deprived mice, exacerbates the vulnerability of dopaminergic neurons, via a dysfunctional astrocyte-microglia-neuron crosstalk [213–215]. Of note, gender differences are also present in the sensitivity to most environmental PD neurotoxin including rotenone and MPTP.

    • Repurposing steroidogenesis inhibitors for the therapy of neuropsychiatric disorders: Promises and caveats

      2019, Neuropharmacology
      Citation Excerpt :

      Although aromatase blockers are well-tolerated, their applicability in neuropsychiatric therapy appears limited, given that the reduction of estrogens is linked to cognitive impairment and greater vulnerability to excitotoxicity (Azcoitia et al., 2001; Garcia-Segura et al., 2003) and risk of neurodegeneration. For example, aromatase inhibition potentiates the nigrostriatal damage after exposure to a dopaminergic neurotoxin in models of Parkinson's disease (Morale et al., 2008). In addition, lack of aromatase in mice has been associated with greater aggression, as well as depressive-like and perseverative behaviors (Hill et al., 2007; Trainor et al., 2006).

    • Aromatase Deficiency and Aromatase Excess

      2014, Genetic Steroid Disorders
    View all citing articles on Scopus
    View full text