7α-Hydroxylation of dehydroepiandrosterone does not interfere with the activation of glucocorticoids by 11β-hydroxysteroid dehydrogenase in EtC cerebellar neurons

https://doi.org/10.1016/j.jsbmb.2013.07.001Get rights and content

Highlights

  • We report that EtC neuronal cerebellar cells possess 11β-HSD1 and H6PDH activity.

  • We examine the interplay of glucocorticoid reduction and DHEA hydroxylation.

  • Glucocorticoid activation by 11β-HSD1 is not affected by 7α-hydroxylation of DHEA.

  • 7α-OH-DHEA is the sole product of DHEA metabolism by EtC neuronal cells.

Abstract

The neuroprotective action of dehydroepiandrosterone (DHEA) in the absence of a known specific receptor has been attributed to its metabolism by different cell types in the brain to various steroids, with a preference to its 7-hydroxylated products. The EtC cerebellar granule cell line converts DHEA almost exclusively to 7α-hydroxy-DHEA (7α-OH-DHEA). It has been postulated that DHEA's 7-OH and 7-oxo metabolites can decrease glucocorticoid levels by an interactive mechanism involving 11β-hydroxysteroid dehydrogenase (11β-HSD). In order to study the relationship of 7-hydroxylation of DHEA and glucocorticoid metabolism in intact brain cells, we examined whether EtC cerebellar neurons, which are avid producers of 7α-OH-DHEA, could also metabolize glucocorticoids. We report that EtC neuronal cells exhibit 11β-HSD1 reductase activity, and are able to convert 11-dehydrocorticosterone into corticosterone, whereas they do not demonstrate 11β-HSD2 dehydrogenase activity. Consequently, EtC cells incubated with DHEA did not yield 7-oxo- or 7β-OH-DHEA. Our findings are supported by the reductive environment of EtC cells through expression of hexose-6-phosphate dehydrogenase (H6PDH), which fosters 11β-HSD1 reductase activity. To further explore the role of 7α-OH-DHEA in EtC neuronal cells, we examined the effect of preventing its formation using the CYP450 inhibitor ketoconazole. Treatment of the cells with this drug decreased the yield of 7α-OH-DHEA by about 75% without the formation of alternate DHEA metabolites, and had minimal effects on glucocorticoid conversion. Likewise, elevated levels of corticosterone, the product of 11β-HSD1, had no effect on the metabolic profile of DHEA. This study shows that in a single population of whole-cells, with a highly reductive environment, 7α-OH-DHEA is unable to block the reducing activity of 11β-HSD1, and that 7-hydroxylation of DHEA does not interfere with the activation of glucocorticoids. Our investigation on the metabolism of DHEA in EtC neuronal cells suggest that other alternate mechanisms must be at play to explain the in vivo anti-glucocorticoid properties of DHEA and its 7-OH-metabolites.

Introduction

It is now well established that dehydroepiandrosterone (DHEA) is produced in the central nervous system independently from its adrenal synthesis [1] and that it has various neuroprotective properties. The many diverse effects of DHEA in the brain, in the absence of a known specific receptor, have been attributed to its metabolic products that are formed by different cell types [2]. For example, in microglia, DHEA is reduced by 17-β-hydroxysteroid dehydrogenase to 5-androstenediol [3], whereas in hippocampal granule cells it is hydroxylated to its 7α form [4]. On the other hand, in astrocytes it is converted to androstenedione (AD) [5], which serves as a precursor to the synthesis of androgens and estrogens; estrogens have been shown to be neuroprotective in hippocampal cells [6], [7] and to enhance learning and memory [rev. by 8]. Collectively, these data suggest the neuroprotection attributed to DHEA is a function of its metabolites, which are intrinsic to distinct cell types.

In hippocampal and cerebellar mouse neuronal cell lines, as well as in rat and mouse brain microsomes, the primary product of DHEA metabolism is 7α-hydroxy-DHEA (7α-OH-DHEA) [4], [9], [10], [11], and to a lesser extent, 7β-OH-DHEA has been reported in rat brain microsomes [12]. In the brain, the conversion of DHEA to 7α-OH-DHEA is primarily attributed to cytochrome P450 CYP7B [13], [14]. The 7-hydroxylated metabolites of DHEA can be neuroprotective [9], [15] and possibly improve memory [16], [17]. Furthermore, in dementia and Alzheimer's disease, levels of CYP7B are lower, which suggests a role for the loss of 7-hydroxylation in these disease states [18].

Although it is important to note that other 7α-hydroxylases are expressed in the brain, i.e. CYP39A1 [19], and liver, i.e. CYP7A [20], thus far, no known P450 enzyme has been identified to be responsible for the conversion of 7α-OH-DHEA to 7β-OH-DHEA [21]. However, it has been shown that 7α-OH-DHEA can be converted to 7β-OH-DHEA through a 7-oxo-DHEA intermediate via 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) [22], [23], the principal activating enzyme of glucocorticoids. This mechanism has yet to be shown to occur in vivo in the brain; additionally, there is no evidence of 7-oxo-DHEA formation in the brain.

The 7α and 7β-OH metabolites of DHEA are known to be more efficacious in their biological actions in various tissues than their parent steroid, DHEA. This is particularly evident in their immunomodulatory activity [10], [24], [25]. Likewise, DHEA and its metabolites have marked anti-stress [26] and anti-glucocorticoid properties [27], suggested to occur via various mechanisms. The inhibition of nuclear translocation of the glucocorticoid receptor has been reported in neurons [28], but not in transfected COS-7 cells [29]. Another proposed mechanism for DHEA's immunomodulatory activity is its involvement in the metabolism of glucocorticoids [rev. by 30]. 11β-HSD1 can convert the inactive glucocorticoid 11-dehydrocorticosterone (11-dehydro-CORT) (cortisone in humans) into corticosterone (CORT) (cortisol in humans) using NADPH as a cofactor; the 11β-HSD2 isoform can perform the reverse reaction using NAD+ as a cofactor [rev. by 31]. In vitro, it has been demonstrated that 7α-OH-DHEA can convert to 7β-OH-DHEA through a 7-oxo-DHEA intermediate via the NADP+ dependent 11β-HSD and this process is reversed by the addition of NADPH [22], [24]. Furthermore, using microsomal fractions, 7OH-DHEA was shown to inhibit the activity of 11β-HSD, therefore blocking formation of CORT. These data are highly suggestive that DHEA's anti-glucocorticoid activity may occur through a putative competitive inhibitory mechanism of 11β-HSD [23], [30]. In the current study, we aimed to determine if the mouse cerebellar neuronal cell line EtC, which is an avid producer of 7OH-DHEA, possesses 11β-HSD activity, and use it as a whole-cell system to investigate the DHEA anti-glucocorticoid paradigm.

Section snippets

Compounds

[1,2,6,7-3H]-labeled DHEA and [3H]-labeled CORT (Perkin Elmer Life Sciences Inc., Waltham, MA, USA) and [3H]-labeled 11-dehydro-CORT (generously donated by the Population Council, The Rockefeller University, New York, NY, USA) were obtained and their purity was determined by thin layer chromatography (TLC) on a silica gel containing a fluorescent indicator on pre-coated aluminum sheets (Fisher, Carlsbad, CA, USA). Unlabeled hormones (7α-OH-DHEA, 7β-OH-DHEA, 7-oxo-DHEA, DHEA, androstenedione,

EtC cells express functional 11β-HSD1

In order to study the relationship of 7-hydroxylation of DHEA and glucocorticoid metabolism in intact brain cells, we examined whether EtC cerebellar neurons, which are avid producers of 7α-OH-DHEA, could also metabolize glucocorticoids. EtC expressed 11β-HSD1 and demonstrated 11β-HSD1 reductase activity by converting [3H]-11-dehydro-CORT into CORT (Fig. 1A and B), but did not express 11β-HSD2 dehydrogenase nor deactivated [3H]-CORT into 11-dehydro-CORT (Fig. 1A and B). 11β-HSD2 expression was

Discussion

In this study, we show that EtC cells possess exclusive 11β-HSD1 reductase activity and convert inactive 11-dehydro-CORT into CORT, consistent with H6GDH expression and lack of 11β-HSD2 isoform expression. As expected, the addition of exogenous 7-oxo-DHEA inhibited the conversion of 11-dehydro-CORT into active CORT. However, EtC cells did not produce their own 7-oxo-DHEA or 7β-OH-DHEA, which are the main inhibitors of 11β-HSD. Furthermore, blocking 7-hydroxylation of DHEA did not increase the

Conflict of interest

The authors have no conflicting financial interests.

Acknowledgements

We would like to thank Dr. Renshan Ge and Chantal M. Sottas (Population Council, Rockefeller University, NY) for generous provision of [3H]-labeled 11-dehydro-CORT, 11β-HSD enzyme inhibitors, and use of the TLC scanning radiometer. This work was supported by the Peter Deane Trust (K.B.).

References (54)

  • B. Robinzon

    Glucocorticoids inhibit interconversion of 7-hydroxy and 7-oxo metabolites of dehydroepiandrosterone: a role for 11beta-hydroxysteroid dehydrogenases?

    Archives of Biochemistry and Biophysics

    (2003)
  • Y. Hu

    Anti-stress effects of dehydroepiandrosterone: protection of rats against repeated immobilization stress-induced weight loss, glucocorticoid receptor production, and lipid peroxidation

    Biochemical Pharmacology

    (2000)
  • C. Muller

    Dehydroepiandrosterone and its 7-hydroxylated metabolites do not interfere with the transactivation and cellular trafficking of the glucocorticoid receptor

    The Journal of Steroid Biochemistry and Molecular Biology

    (2004)
  • C. Muller et al.

    The native anti-glucocorticoid paradigm

    The Journal of Steroid Biochemistry and Molecular Biology

    (2006)
  • S.A. Latif

    Endogenous selective inhibitors of 11beta-hydroxysteroid dehydrogenase isoforms 1 and 2 of adrenal origin

    Molecular and Cellular Endocrinology

    (2005)
  • K. Bulloch et al.

    A new method for the establishment of neuronal cell lines from the mouse brain

    Life Sciences

    (1978)
  • K. Bulloch et al.

    The derivation and characterization of neuronal cell lines from rat and mouse brain

    Brain Research

    (1977)
  • A.K. Agarwal

    Cloning and expression of rat cDNA encoding corticosteroid 11 beta-dehydrogenase

    The Journal of Biological Chemistry

    (1989)
  • L.K. Klaidman et al.

    High-performance liquid chromatography analysis of oxidized and reduced pyridine dinucleotides in specific brain regions

    Analytical Biochemistry

    (1995)
  • M.A. Pelissier

    Protection against dextran sodium sulfate-induced colitis by dehydroepiandrosterone and 7alpha-hydroxy-dehydroepiandrosterone in the rat

    Steroids

    (2006)
  • M.A. Pelissier

    Antioxidant effects of dehydroepiandrosterone and 7alpha-hydroxy-dehydroepiandrosterone in the rat colon, intestine and liver

    Steroids

    (2004)
  • D.K. Fukushima

    Studies in steroid metabolism: XXV. Isolation and characterization of new urinary steroids

    The Journal of Biological Chemistry

    (1954)
  • A. Gottfried-Blackmore

    Characterization of a cerebellar granule progenitor cell line, EtC.1, and its responsiveness to 17-beta-estradiol

    Brain Research

    (2007)
  • P. Robel et al.

    Dehydroepiandrosterone (DHEA) is a neuroactive neurosteroid

    Annals of the New York Academy of Sciences

    (1995)
  • I.H. Zwain et al.

    Dehydroepiandrosterone: biosynthesis and metabolism in the brain

    Endocrinology

    (1999)
  • Y. Akwa

    Astrocytes and neurosteroids: metabolism of pregnenolone and dehydroepiandrosterone: regulation by cell density

    The Journal of Cell Biology

    (1993)
  • Y. Goodman

    Estrogens attenuate and corticosterone exacerbates excitotoxicity, oxidative injury, and amyloid beta-peptide toxicity in hippocampal neurons

    Journal of Neurochemistry

    (1996)
  • Cited by (8)

    • DHEA-induced modulation of renal gluconeogenesis, insulin sensitivity and plasma lipid profile in the control- and dexamethasone-treated rabbits. Metabolic studies

      2016, Biochimie
      Citation Excerpt :

      Regarding mechanisms by which DHEA may reverse insulin resistance, there are several possibilities: 1) a decrease in both expression and activity of 11β-HSD1 in liver and adipocytes [9,72], 2) activation of PPARα, which binds DHEA, leading to fat oxidation, repression of its synthesis, and in consequence a decreased fat deposition in liver and muscle [68], 3) a decline of circulating levels of the inflammatory cytokines [66,68], 4) a decrease of leptin expression in adipocytes [72], 5) modulation of transcriptional GR activity [8], 6) an increased AMPK activity [73], and 7) an enhancement of insulin action in muscle by improving an impaired activation of both phosphatidyl inositol 3-kinase and Akt/PkB [68]. The mechanism by which DHEA exerts its pleiotropic effects in humans and rodents may also involve the action of its more active metabolites rather than effect of DHEA alone [42]. DHEA hydroxylation (catalyzed by cytochromes P450) has been reported to occur in a wide variety of tissues and organs, like: muscle, brain, spleen, thymus, skin, intestine, colon [74] and liver [75].

    • Determination of seven selected neuro- and immunomodulatory steroids in human cerebrospinal fluid and plasma using LC-MS/MS

      2015, Steroids
      Citation Excerpt :

      In the 1990s it was reported that endogenous 7-oxo-metabolite of DHEA could act as an ergosteroid, enhancing the activity of several enzymes that influence a thermogenic system in rat liver [15,16]. Later also the neuroprotective and antiglucocorticoid actions of 7-oxo-DHEA were discovered [1,17,18]. The abovementioned 7-oxygenated steroids are metabolized by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1): 7α-OH-DHEA is a substrate for the 11β-HSD1, which converts 7α-OH-DHEA into 7β-OH-DHEA via 7-oxo-DHEA and also vice versa [19–21].

    • Steroid hormones in prediction of normal pressure hydrocephalus

      2015, Journal of Steroid Biochemistry and Molecular Biology
      Citation Excerpt :

      Recent findings using rat cerebellar granule cell line however did not confirmed this mechanism. The discrepancy could be due to reductive environment of the cell system used, in which hexose-6-phosphate dehydrogenase is the major player [69]. Taking into account the reduced CSF DHEA metabolites in NPH and the opposite trend observed in AD and VD [66–68], the CSF DHEA metabolic profiling may successfully differentiate NPH patients from those with AD or VD.

    View all citing articles on Scopus
    1

    These authors contributed equally to this manuscript.

    View full text