Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Steroids 77 (2012) 10–26

Contents lists available at SciVerse ScienceDirect

Steroids
journal homepage: www.elsevier.com/locate/steroids

Review

Oxidative metabolism of dehydroepiandrosterone (DHEA) and biologically


active oxygenated metabolites of DHEA and epiandrosterone (EpiA) – Recent reports
Laïla El Kihel ⇑
Université de Caen Basse-Normandie, U.F.R. des Sciences Pharmaceutiques, Centre d’Etudes et de Recherche sur le Médicament de Normandie, UPRES EA-4258,
FR CNRS INC3M, Boulevard Becquerel, 14032 Caen Cedex, France

a r t i c l e i n f o a b s t r a c t

Article history: Dehydroepiandrosterone (DHEA) is a multifunctional steroid with a broad range of biological effects in
Received 21 April 2011 humans and animals. DHEA can be converted to multiple oxygenated metabolites in the brain and
Received in revised form 14 September peripheral tissues. The mechanisms by which DHEA exerts its effects are not well understood. However,
2011
evidence that the effects of DHEA are mediated by its oxygenated metabolites has accumulated.
Accepted 18 September 2011
Available online 2 October 2011
This paper will review the panel of oxygenated DHEA metabolites (7, 16 and 17-hydroxylated deriva-
tives) including a number of 5a-androstane derivatives, such as epiandrosterone (EpiA) metabolites. The
most important aspects of the oxidative metabolism of DHEA in the liver, intestine and brain are
Keywords:
Oxygenated DHEA metabolites
described. Then, this article reviews the reported biological effects of oxygenated DHEA metabolites from
Epiandrosterone recent findings with a specific focus on cancer, inflammatory and immune processes, osteoporosis, ther-
Oxidative DHEA metabolism mogenesis, adipogenesis, the cardiovascular system, the brain and the estrogen and androgen receptors.
Hydroxy-DHEA Ó 2011 Elsevier Inc. All rights reserved.
Hydroxy-EpiA
DHEA metabolites activities

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
2. Oxidative metabolism of DHEA and epiandrosterone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.1. Metabolism in the liver. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.1.1. DHEA metabolites. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.2. Epiandrosterone metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.2. Metabolism in the intestine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.2.1. DHEA metabolites. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.2.2. EpiA metabolites. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.3. Metabolism in the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.4. DHEA metabolites in cerebrospinal fluid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3. Biological activities of DHEA and EpiA metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

Abbreviations: AD, Alzheimer’s disease; DHEA, 3b-hydroxy-androst-5-en-17-


one, dehydroepiandrosterone; EpiA, 3b-hydroxy-5a-androstan-17-one, epiandros-
terone; GC–MS, gas chromatography–mass spectrometry; LC–MS, liquid chroma-
tography–mass spectrometry; 3b,17b-AAD, 5a-androstane-3b,17b-diol; 3b,7a,17b-
AAT, 5a-androstane-3b,7a,17b-triol; 3b,7b,17b-AAT, 5a-androstane-3b,7b,17b-
triol; ADT, 5a-3b-hydroxy-androstan-17-one, androsterone; Adione, 5a-andro-
stane-3,17-dione; 4-adione, androst-4-ene-3,17-dione; 7a-HO-DHEA, 7a-hydroxy-
DHEA; 7b-HO-DHEA, 7b-hydroxy-DHEA; 3b,17a-AED, androst-5-ene 3b,17a diol;
3b,17b-AED, androst-5-ene-3b,17b-diol; 3b,7a,17b-AET, 5a-androst-5-ene-
3b,7a,17b-triol; 3b7b,17b-AET, 5a-androst-5ene-3b,7b,17b-triol; T, testosterone.
⇑ Tel.: +33 0 2 31 56 68 12; fax: +33 0 2 31 56 68 03.
E-mail address: laila.elkihel@unicaen.fr

0039-128X/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.steroids.2011.09.008
L.E. Kihel / Steroids 77 (2012) 10–26 11

3.1. DHEA metabolites and anticancer properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18


3.2. DHEA metabolites and their effects on the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3.2.1. DHEA metabolites and neuroprotective effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3.2.2. DHEA metabolites and memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3.2.3. DHEA metabolites and neurodegenerative diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3.3. DHEA metabolite effects on inflammatory and immune processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.4. DHEA metabolites and osteoporosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.5. DHEA metabolites and thermogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.6. DHEA metabolites and adipogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.7. DHEA metabolites and the skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.8. DHEA metabolites and the cardiovascular system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.9. DHEA metabolites and estrogenic receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.10. DHEA metabolites and androgenic receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
3.11. DHEA metabolites and the human prostate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

1. Introduction Clinical and experimental studies have shown that DHEA/


DHEAS in humans and other mammals are multi-functional
Dehydroepiandrosterone (3b-hydroxy-androst-5en-17-one, steroids implicated in a broad range of biological processes, includ-
DHEA) is an endogenous steroid synthesized in the adrenal cortex, ing obesity [4], diabetes [5], bone metabolism [6], neuroprotection
gonads, brain, and gastrointestinal tract. Its sulfated derivative, [7], atherosclerosis [8], and anti-tumorigenesis [9]. DHEA and its
DHEAS, is the most abundant circulating steroid in young adult sulfate are also potent neuroactive steroids through their modula-
humans. Its levels decline dramatically with age and have been tory effects on the c-aminobutyric (GABA) and N-methyl-D-aspar-
correlated with degenerative changes related to aging. DHEA may tate (NMDA) receptors [10].
directly maintain bone, muscle, skin, and brain cells, modify the Moreover, DHEA is the physiological precursor in the synthesis
immune response, inflammation, and lipid metabolism, and of androgens and estrogen via androst-4-ene-3,17-dione (4-adi-
ameliorate the deleterious effects of glucocorticoids [1–3]. one) in humans [11,12]. Many studies have been undertaken in

Oxygenated DHEA metabolites

O
O O

O O
HO HO S
HO -O
Pregnenolone O DHEA-sulfate
Cholesterol
Dehydroepiandrosterone (DHEA)
Other steroids
O
OH OH O

O
O O HO
H Androst-4-ene-3,17-dione (4-Adione) H
Dihydrotestosterone Testosterone
Etiocholanolone

OH
O hydroxylated 4-Adione metabolites

oxygenated Epi-A metabolites


HO HO
H H
5α-androstane-3β,17β−diol (3β,17β-AAD)
Epiandrosterone (EpiA)
Fig. 1. Biosynthetic pathway of DHEA and EpiA modified scheme from [21,22].
12 L.E. Kihel / Steroids 77 (2012) 10–26

O
OH

O
O O
OH S HO
-O O
DHEA-sulfate OH
Androst-5-ene-3β,17β-diol
HO
3β,16α-dihydroxy-androst-5-ene-17-one
O

O
O
OH O
HO
OH
O
Androst-4-ene-3,17-dione
HO O
HO
DHEA

O OH OH
O

O O
O O HO OH HO OH S OH
OH
-O
S
7α−ΟΗ−DHEA -O O
O

O O OH OH

O O
O O HO O O
O HO O S
S
-O O
7- oxo-DHEA -O O

O O OH OH

O O O
O HO OH HO OH OH
S OH S
-O O 7β−ΟΗ−DHEA -O O

Fig. 2. An overview of summary oxygenated DHEA metabolites identified after incubation of DHEA with rat liver homogenate Scheme slightly modified from Marwah et al.
[14]. See also [21,27,29].

attempts to elucidate the mechanism(s) responsible for these ef- 3b-hydroxysteroids include pregnenolone and 5a-androstane-
fects, but the data available so far indicate that DHEA acts at vari- 3a,17-a-diol in tissues, such as the brain, liver, skin, and joints,
ous sites and levels rather than by a unifying mechanism. The where the hydroxylase is expressed. These tissues also contain
mechanism by which DHEA exerts its pleiotropic effects in humans NADP(H)-dependent 11b-hydroxysteroid dehydrogenase type 1
and rodents is not well understood but may involve metabolism of (11b-HSD1), which converts 7a-hydroxysteroids to the corre-
DHEA into biologically active oxygenated metabolites [13,14]. sponding 7b-hydroxysteroids [15–19]. Thus, DHEA metabolism is
the key to understanding several physiological events. DHEA
2. Oxidative metabolism of DHEA and epiandrosterone metabolism has also been studied in the brain, spleen, thymus,
perianal skin, ventral skin, intestine, colon, and muscle tissues
DHEA is a naturally occurring C-19 adrenal steroid that is from mice [20].
derived from cholesterol by a series of cytochrome P450 reactions, Herein, we focus on the metabolism of DHEA and EpiA in the li-
and DHEA is a precursor for the metabolite epiandrosterone (3a- ver, intestine, and brain.
hydroxy-5a-androstan-17-one, EpiA). It should be noted that EpiA
is formed by two mechanisms: (1) total enone ring A reduction of 2.1. Metabolism in the liver
androst-4-ene-3,17-dione (4-adione) and (2) selective reduction of
the D4 double bond and 3a-ketone of 4-adione to generate 5a- The liver is one of the most important organs in the metabolism
androstane-3a,17a-diol (3a,17a-AAD), followed by 17a-oxidation of drugs [23]. Biotransformation involves phase I and phase II path-
to generate EpiA (Fig. 1). DHEA is metabolized to androgens and ways and mainly takes place in the liver and intestine [24,25].
estrogens in steroidogenic tissues. It is also metabolized with EpiA Phase I is usually oxidative, and these reactions are mainly cata-
into oxygenated derivatives in mammalian tissues, such as the lyzed by various enzymes of the Cytochrome P450 (CYP) supergene
intestine, brain, and especially human liver. The cytochrome family, which are predominantly expressed in the liver and play a
P450-7B1 (CYP7B1) uses NADPH and is responsible for the 7a- central role in drug metabolism. In phase II, such oxidized moieties
hydroxylation of all 3b-hydroxysteroids (except cholesterol). The are conjugated with highly polar molecules, mainly glucuronic acid
L.E. Kihel / Steroids 77 (2012) 10–26 13

or sulfate [26]. When these steroids are administered per os to gas chromatography–mass spectrometry (GC–Ms) analysis). In
humans, the first organ encountered is the liver, where extensive adult liver microsomes, CYP3A4 and CYP3A5 were the P450 cyto-
metabolism takes place. Most orally administrated steroids are chromes responsible for the production of 7a-hydroxy-DHEA,
metabolized in the liver and excreted in the urine. As the liver is 16a-hydroxy-DHEA, and the previously unidentified hydroxylated
responsible for xenobiotic (including steroids) elimination through metabolite 7b-hydroxy-DHEA (reported by Fitzpatrick et al. [13]).
phase I (Cytochrome P450-mediated hydroxylation) and phase II Whereas the fetal/neonatal form CYP3A7 produced 16a-hydroxy
(conjugation) processes, it has been suggested that hydroxylated and 7b-hydroxy-DHEA, CYP3A23 uniquely generated 7a-hydro-
steroid derivatives mainly protect against excessive steroid xy-DHEA. Furthermore, other P450s, including CYP2B1, CYP2C11,
production. and CYP2D1, are responsible for 16a-hydroxy-DHEA metabolite
production in rat liver microsomal fractions [36]. Additionally, Ste-
2.1.1. DHEA metabolites vens et al. observed that CYP3A7 is expressed nearly exclusively in
DHEA metabolites have been studied extensively [14,27], and human neonates [28]. The results of these studies demonstrate
many investigators have investigated the conversion of DHEA to that the regio- and stereo-selectivity of hydroxylation of DHEA
a variety of metabolites using a specific tissue fraction as the en- by specific P450s account for the unique DHEA metabolite profiles
zyme source [28–33]. Special attention has been paid to the study formed by various species.
of oxygenated DHEA metabolites [14]. The transformation of DHEA A recent report attempted to study the transformation of DHEA
was examined in whole rat liver homogenate in the presence of and its 7- or 16a-hydroxylated derivatives in S9 fractions of human
malic acid using NADH and NADPH. The aim was to predict the liver microsomes with the cofactor NAD(H) or NADP(H) using a
type of reactions that might occur in vivo, where the product of radiolabeled steroid substrate for quantification and gas chroma-
one organelle may be the substrate for another, and to study the tography–mass spectrometry (GC–MS) for identification [15]. The
timing of metabolite appearances to predict the sequence of their results showed that DHEA was transformed into its 7- or 16a-
formation. Twenty oxygenated DHEA metabolites were identified hydroxylated metabolites and 17b-hydroxylated derivatives in
and characterized using the highly specific and selective analysis the presence of NAD(P)H. 17b-Hydroxysteroid dehydrogenase
method of liquid chromatography–mass spectrometry (LC–MS). was responsible for the reduction of 17-oxo-steroids into 17b-
These results showed that 7a-hydroxy-DHEA was the first product hydroxysteroids. The generation of 16a-hydroxy-DHEA from DHEA
formed from DHEA, which was converted via the sequence 7a-hy- indicated the expression of CYP3A4 and CYP3A5 in liver S9 frac-
droxy-DHEA ? 7-oxo-DHEA ? 7a-hydroxy-DHEA. These deriva- tions. It was demonstrated that liver microsomal preparations
tives were also converted to diols, triols and sulfate esters (Fig. 2). produce equal amounts of 7-oxo and 7b-hydroxy-DHEA from
In the livers of rats, mice, and humans, 7a-hydroxylation of 7b-hydroxy-DHEA. It was also demonstrated that 7a/7b-hydroxy-
DHEA is carried out by several P450 cytochromes (such as CYP7B1). and 7-oxo-DHEA are not substrates for 16a-hydroxylase. In
Additionally, one of the most abundant cytochromes in the liver is addition, these data confirm the 7a/7b interconversion via a 7-
the CYP3A forms CYP3A4 and CYP3A5, which have been reported oxo-DHEA intermediate using an oxidoreduction process catalyzed
to carry out the 7a-hydroxylation of DHEA [13]. Fitzpatrick et al. by 11a-hydroxysteroid dehydrogenase type 1. However, 17a-
identified the DHEA metabolites formed by rodent and human liver hydroxysteroid dehydrogenase (17a-HSD) is responsible for the
microsomal fractions in the presence of NADPH and O2. Hydroxyl- reduction of 17-oxo-steroids into 17a-hydroxysteroids [37]. These
ated metabolites, principally 7a-HO-DHEA, 7-oxo-DHEA, and 16a- authors have shown that 17a-HSD converts DHEA to androst-5-
HO-DHEA, were identified in both species [13]. These authors and ene-3a,17a-diol (3a,17a-AED) (Fig. 3). To date, little information
others have demonstrated the conversion of 7a- and 7b-HO-DHEA is available about the biosynthesis, distribution, and biological
to 7-oxo-DHEA by rat and human isozymes of the 11b-hydroxy- function of 3a,17a-AED.
steroid dehydrogenases [34,35]. 11b-HSD1 has been reported to catalyze the interconversion of
Another study identified and quantitated the liver microsomal several 7-oxygenated sterols and steroids. Muller et al. used micro-
oxidative metabolism of DHEA by rodent, hamster, pig, and human somes from yeast expressing recombinant human 11b-HSD1 and
microsomal fractions (quantification of DHEA and metabolites by demonstrated the interconversion between 7-hydroxy- and

O
OH

17α-HSD
HO
HO
DHEA Androst-5-ene-3β,17α-diol (3β,17α−AED)

3β-HSD
3β-HSD

O OH

17α-HSD
O O
Androst-4-ene-3,17-dione (4-Adione) 17α,-hydroxy-androst-4-en-3-one (Epi-Testosterone)

Fig. 3. Conversion of DHEA into Androst-5-ene-3b,17a-diol (3b,17a-AED) by the enzyme 17a-hydroxysteroid dehydrogenase (17a-HSD) [37].
14 L.E. Kihel / Steroids 77 (2012) 10–26

Androst-5-ene-3β,7α,16α,17β-tetrol Androst-5-ene-3α,7α,16α,17β-tetrol
OH OH OH OH

OH OH OH OH

I II III IV
HO OH HO OH HO OH HO OH

Androst-5-ene-3α,7β,16α,17β-tetrol
Androst-5-ene-3β,7β,16α,17β-tetrol
Fig. 4. Androst-5-enetetrols obtained by human liver microsomes identified by Ahlem et al. [39].

OH

HO
H

O O
O
OH
OH

HO HO
HO OH H H
HO OH H Epiandrosterone (EpiA)
H
7α-HO-EpiA
O
?
?
O O

O
? H

HO O HO OH
H H

7-Oxo-EpiA 7β-HO-EpiA
Not identified

Fig. 5. Summary EpiA oxygenated metabolites identified after EpiA incubation with human liver S9 fractions according to Chalbot et al. [15]. See also [21]. ?: Enzyme
pathways are not clearly identified.

7-oxo-DHEA. A high level of production of 7b-hydroxy-DHEA was 7-oxo- and 7-hydroxy-DHEA is regulated by 11bHSD1 and greatly
observed. The Km values obtained for the reduction of 7-oxo-DHEA depends on the co-expression of hexose-6-phosphate dehydroge-
and the oxidation of 7b-hydroxy-DHEA were about half that for nase [32]. These findings identified 11b-HSD1 as the enzyme
cortisone and cortisol, respectively, indicating that 11b-HSD1 has responsible for the generation of various 7b-hydroxysteroid
a higher affinity for the 7-oxo-DHEA metabolites than for glucocor- metabolites [38]. Novel components of the human metabolome
ticoids [19]. were obtained by metabolic conversion of androst-5-ene precur-
Recent findings suggest preferential formation of 7b-hydroxy- sors to tetrols (I–IV) by human liver microsomes (Fig. 4). Of these
DHEA from 7-oxo-DHEA as well as some conversion of tetrols, the 7b-hydroxy epimers (I and III), but not the 7a-epimers,
7a-hydroxy-DHEA to 7b-hydroxy-DHEA. The equilibrium between were detected in human plasma and urine. This is the first report of

O OH

17β-hydroxysteroid oxido-reductase

HO R HO R

R = H, DHEA R = H, 3β-17β-AED
R = α-OH, 3β,7α,17β-AET
R = α-OH, 7α-HO-DHEA
R = β-OH, 3β,7β,17β-AET
R = β-OH, 7β-HO-DHEA
Fig. 6. Transformation of DHEA, 7a-hydroxy-DHEA and 7b-hydroxy-DHEA substrates in microsomes of human intestine. The steroid metabolites were identified by GC/MS
analysis. Scheme modified from Chalbot et al. [47].
L.E. Kihel / Steroids 77 (2012) 10–26 15

a natural source of the androst-5-enetetrols 7b-epimers in human However, to the best of our knowledge, very few studies on
tissues [39]. DHEA, EpiA, and their metabolites have been carried out in the
intestine.

2.1.2. Epiandrosterone metabolites


2.2.1. DHEA metabolites
Androgenic steroids are derived from DHEA after puberty and
In one study, the [4–14C]-labeled steroid substrates (DHEA, 7a-
include epiandrosterone, which shares a closely related structure
hydroxy-DHEA and 7b-hydroxy-DHEA) were produced [46] by
with DHEA and is 7a-hydroxylated [16]. The 7a-hydroxylated
incubation of different cofactors with microsomes of human intes-
derivatives of EpiA may provide inactivation of androgen potencies
tine, and the metabolites were identified by gas chromatography-
and are of interest because of their promising biological effects
mass spectrometry [47]. The sole metabolites resulting from the
[40,41]. 7a-Hydroxy-EpiA is produced from EpiA via cytochrome
DHEA incubations were 17b-reduced derivatives, obtained by
P450-7B1 (CYP7B1) in tissues where the hydroxylase is expressed,
reduction of 17-ketone steroids. This enzymatic reduction process
including the brain, liver, skin and joints [16,18,42]. These tissues
implies the presence of a 17b-hydroxysteroid oxidoreductase. The
also contain NADP(H)-dependent 11b-HSD1, which converts 7a-
same finding applies to 7a-hydroxy-DHEA and 7b-hydroxy-DHEA
hydroxysteroids to the corresponding 7b-hydroxysteroids
metabolism, where 17b-reduction resulted in the respective triol
[19,42,43].
production (Fig. 6).
Investigations of EpiA metabolism in S9 fractions of the human
Except for 17b-reduced transformation, no other metabolites of
liver provided evidence for EpiA transformation to their 17b-
DHEA, 7a-hydroxy-DHEA, and 7b-hydroxy-DHEA have been ob-
hydroxylated derivatives, 7- or 16-a-hydroxylated metabolites
served. However, the formation of 17b-hydroxy-metabolites pro-
(Fig. 5). Provided that 16a-hydroxy-EpiA is generated by cyto-
duced by either the 17b-hydroxysteroid oxidoreductase or by
chrome P450 CYP3A4, a significantly larger amount of 16a-EpiA
other enzymes cannot be directly identified. These results also
was obtained in females than in males. 7a-Hydroxy-EpiA and 7b-
show that the tested intestine microsome fraction did not contain
hydroxy-EpiA interconvert to a degree, but no 7-oxo-EpiA interme-
the CYP7B1 responsible for the 7a-hydroxylation of DHEA. Neither
diate has been obtained, in contrast to the 7-hydroxylated DHEA
oxidation products of 7a- and 7b-hydroxy-DHEA nor inter-conver-
inter-conversion via 7-oxo-DHEA [15]. It can be postulated that
sion of these steroids were detected in the digests.
an unknown epimerase is involved in the inter-conversion process.
These data provided evidence that 11b-HSD1, depending on the
2.2.2. EpiA metabolites
NAD(H) or NADP(H) cofactors, can function as an epimerase that
Metabolism of EpiA, 7a-hydroxy-EpiA and 7b-hydroxy-EpiA
catalyzes the interconversion between 7a- and 7b-hydroxy-EpiA
was explored in the same manner by Chalbot and Morfin [47].
and between 5a-androstane-3b,7a,17b-triol (3b,7a,17b-AAT) and
The major metabolites of DHEA obtained were 17b-reduced deriv-
5a-androstane-3b,7b,17b-triol (3b,7b,17b-AAT), respectively, with
atives, and a 7a-hydroxylated metabolite of EpiA was not found
a preferential formation of the 7b-hydroxy metabolite [42,43].
(Fig. 7).
Additionally, no 5a,7-oxo-androstan-3b,17b-diol intermediate
In contrast, experiments with 7a-hydroxy-EpiA or 7b-hydroxy-
was present in the 5a-androstan-3b,7a,17b-triol/5a-androstan-
EpiA have provided evidence for the inter-conversion of these
3b,7b,17b-triol inter-conversion with a yeast-expressed
steroids. No trace of a 7-oxo derivative in this epimerization was
recombinant human 11bHSD1 [43]. Instead, interconversion of
detected. The identity of the enzyme responsible for the 7a-hydro-
the 7a- and 7b-hydroxylated forms was observed, with a prefer-
xy-EpiA/7b-hydroxy-EpiA inter-conversion is unknown, and to our
ence for the production of the 7b-hydroxylated epimers. Also, EpiA
knowledge, few studies have been carried out to investigate
was oxidized at the 3b-position; this is in contrast with DHEA.
whether an oxidoreductive process is used in this inter-conversion
Additional recent studies have not clearly indicated the presence
[21,42]. This is in contrast with results on 7a- and 7b-hydroxy-
of 7-oxo-EpiA as an intermediate in the conversion of 7a-EpiA to
DHEA, where no inter-conversion was observed. This makes the
7a-EpiA [21].
presence of an 11b-HSD1 in the human intestine doubtful.
However, it is possible that human intestine microsomes used
2.2. Metabolism in the intestine 7a- and 7b-hydroxy-EpiA and not the 7-hydroxy-DHEA epimers
as substrates for an unknown epimerase [47].
DHEA metabolism has been widely studied in the intestine and
other tissues (brain, spleen, thymus, skin, colon, coecum and mus- 2.3. Metabolism in the brain
cle) from mice. 7a-Hydroxy-DHEA and androst-5-ene-3b,17b-diol
were identified as DHEA metabolites in digests of all tissues DHEA and pregnenolone are formed in the brain independent of
[20,44,45]. the adrenal glands and gonads and are called neuro-steroids. While

O OH

17β-hydroxysteroid oxido-reductase

HO
H R HO R
H

R = H, EpiA R = H, 3β,17β-AAD
R = α-OH, 7α-HO-EpiA R = α-OH, 3β,7α,17β−ΑΑΤ
R = β-OH, 7β-HO-EpiA R = β-OH, 3β,7β,17β-AAT
Fig. 7. Transformation of EpiA, 7a-hydroxy-EpiA or 7b-hydroxy-EpiA substrates in microsomes of human intestine. The steroid metabolites were identified by GC/MS
analysis. Scheme modified from Chalbot et al. [47].
16 L.E. Kihel / Steroids 77 (2012) 10–26

DHEA is present at high levels in the blood under normal condi- 7a-HO-DHEA and 3b,17b-AED in the aging brains of healthy hu-
tions, it is found in the brain at even higher levels than in the man subjects. The structures of the brain that were studied were
periphery. the frontal cortex, hippocampus, amygdala, cerebellum and stria-
The first step in steroid biosynthesis corresponds to the forma- tum. The synthesis of 7a-HO-DHEA was found in all of the brain re-
tion of pregnenolone from available cholesterol by oligodendro- gions evaluated. The amount of 7a-HO-DHEA formed in the frontal
cytes in the rat brain, which are then converted to DHEA by the cortex was significantly higher compared with that found in all of
cytochrome P450 enzyme 17a-hydroxylase (CYP17). This enzyme, the other brain regions [61]. The synthesis of 3b,17b-AED was also
which is characteristic of all steroidogenic cells, has been shown to observed in all the human brain structures studied. However, a sig-
be expressed in the nervous system of different classes of verte- nificant difference was observed between the regions. The cerebel-
brates, including amphibians [48,49], birds [50], and mammals lum and striatum had significantly higher conversion rates of
[51–53]. However, the expression of this enzyme and its activity DHEA to 3b,17b-AED compared to the frontal cortex, hippocampus
are undetectable in the adult brain [54]. A recent study showed and amygdala. The formation of 3b,17b-AED from DHEA in the ro-
that DHEA formation is independent of cytochrome P450 17a- dent brain has also been documented [57]. 7a-HO-DHEA, 7b-HO-
hydroxylase/C17,20-lyase activity in the mouse brain. This is the DHEA, and 16a-HO-DHEA are derived from DHEA in the human
first in vivo report demonstrating that there is a DHEA biosynthetic brain, where 7a-hydroxylation and 16a-hydroxylation are cata-
pathway in brain tissue that is distinct from the CYP17-dependent lyzed by CYP7B1 and CYP3A7, respectively [62,63]. The 5-andos-
pathway present in peripheral steroidogenic tissues [55]. To date, tenediols (3a17a-AED and 3a, 17b-AED) are produced by the
this controversy remains unresolved. metabolism of DHEA by 17a-hydroxysteroid dehydrogenase or
DHEA may be metabolized to active metabolites in the brain, 17b-hydroxysteroid dehydrogenase, respectively [37,64–66].
which is then responsible for some or all of the neuroprotective ef- 3b,17a-AED and 3b,17b-AED are neuro-steroids produced in neu-
fects. Although the metabolic profile of DHEA in the brain has not ro-ectodermal tissue by the metabolism of DHEA, and they exist
been fully elucidated, 7a-HO-DHEA and 7b-HO-DHEA are the most in the a- and b-epimeric forms [67]. It has been suggested that
abundant metabolites, and they play important roles in the central 3b,17b-AED and 17b-hydroxysteroid dehydrogenase, which are
nervous system [56]. mainly expressed in astrocytes and microglia, may play a neuro-
Numerous studies have demonstrated that the adult rodent protective role in the brain [68,69]. 17a-Hydroxysteroid dehydro-
brain is the site of extensive conversion of DHEA to 7a-hydroxy- genase is also expressed in the central nervous system, as well as
DHEA [56,57]. However, the metabolism of neuro-steroids, includ- other organs, but the biological function of 3b,17a-AED has not
ing DHEA, in the adult human brain has been not extensively stud- been clearly identified [37,68].
ied [58,59]. A recent study reported a detailed analysis of steroids in the
Cytochrome P4507B is principally expressed in the brain adult male rat brain using GC–MS and applying the identification
[13,60] and has been found to be responsible for the 7a-hydroxyl- criteria of retention time and diagnostic ion ratios at a high level
ation of several 3b-hydroxysteroids, including DHEA, and EpiA. of stringency. Among the steroids identified were DHEA and EpiA,
One study describes the in vitro conversion of DHEA into both which was detected in two-ion Selected Ion Monitoring (SIM, in

O
OH
O O
OH S
-O O
DHEA-sulfate

HO
3β,16α-dihydroxyandrost-5-ene-17-one HO
Androst-5-ene-3β,17β-diol

O
O

HO OH
7α−ΗΟ−DHEA
HO
DHEA ?
?
?
OH O
O

?
HO HO O
Androst-5-ene-3β,17α-diol 7- oxo-DHEA
HO OH
not identified not identified
7β−ΗΟ−DHEA

Fig. 8. Summary oxygenated DHEA metabolites identified in the brain. ?: Enzyme pathways are not clearly identified.
L.E. Kihel / Steroids 77 (2012) 10–26 17

Table 1
Focus on recent finding of oxygenated DHEA metabolite activities.

DHEA metabolites Biological effect References


7oxo-DHEA, 7a-HO-DHEA, 7b-HO-DHEA, Anticancer, cell lines Hep G2, Caco-2, HT-29 [29]
Androsterone, EpiA, Etiocholanolone
3b,17a-AED, 3b,17b-AED Anticancer cell lines: Basal growth of murine RAW 264.7, P388D1, and human HL-60 cells, human [83–
U937 lyphomaT98G and U251 MG gioblastoma, T98G, U87MG, U251MG, LN-18, LN-229 and LN-Z308 85,87]
glioma cell lines
3b,17a-AED, 3b,7a,17a-AET, 3b, 7b,17a-AET, induced apoptosis in U937 lymphoma cells and autophagy in T98G [88]
3b,7a,17b-AET
Effects on the brain
7-oxo-DHEA acetate Beneficial effect for memory in old mice by antagonizing the GABAA receptor [96]
7a-HO-EpiA and 7b-HO-EpiA, 7b-HO-EpiA protection against hypoxia-induced neuronal damage, neuroprotective in two in vivo rat models of [40]
cerebral ischaemia-induced neurodegeneration
7b-HO-EpiA neuroprotective effect in animal models of Alzheimer’s disease [41]
Effects in inflammatory and in immune process
7a-HO-DHEA rheumatoid arthritis: 7a-OH-DHEA levels is 5-fold higher than nonarthritic mice [116–118]
7b-HO-EpiA Anti-oxidant and anti-inflammatory: reduced prostaglandin E2 (PGE2) production [112]
7a-HO-DHEA High antioxidant effect in the colon than DHEA, protective effect against sodium dextran sulfate- [110,111]
induced colitis in rats
7b-HO-EpiA regulation of cytoprotective prostaglandins from human mononuclear cells [113]
7a-HO-DHEA, 7b-HO-DHEA and 7b-HO-EpiA Regulation of the prostaglandin synthesis pathway in human peripheral blood monocytes [114]
3b, 7b,17b-AET limit the production of autoimmune Th1 associated cytokines, and ultimately may be beneficial for [120]
patients with multiple sclerosis or other autoimmune diseases
3b, 7b,17b-AET improves survival in a rodent model of traumatic shock [122]
Androst-5-ene-3a,7b,16a,17b-tetrol, Androst-5- Anti-inflammatory effects in rodent disease models of multiple sclerosis, lung injury, chronic [39]
ene-3b,7b,16a,17b-tetrol prostatitis, and colitis
Osteoporosis
3b,17a-AED, 3b,17b-AED, 3b,7b,17b-AET, 3b,17a-AED promotes bone resorption while 3b,17a-AED inhibits it [132]
3b,7b,17b-AET
3b, 7b,17b-AET preserves bone mineral content, limits cortical cancellous bone loss and preserves endochondral [134]
growth rate
Thermogenesis
7-oxo-DHEA Effect on temporarily actual levels of thyroid hormones [143]
7-oxo-DHEA and 7-oxo-DHEA acetate Thermomodulating agent [137,142]
7a- and 7b-HO-DHEA 7b-HO-DHEA levels, decreased in patients with manifest hypothyroidism (negative relationship) [144]
Adipogenesis
DHEA, 7-oxo-DHEA, 7b-HO-DHEA, 7b-HO- DHEA act as a thermogenic agent in adipocytes, but 7-oxo-DHEA appears to act as a nonthermogenic [147]
DHEA, 3b, 7a,17b-AET, 3b, 7b,17b-AET enhancer of differentiation
Cardiovascular system
3b,17b-AED improves cardiovascular function and organ blood flow [159,160]

mass spectrometry) of the sulfate fraction but not confirmed with neuro-steroid levels in humans. Recent findings indicate that the
three-ion SIM. This inconsistency is probably related to the analyt- CSF level of DHEA is positively correlated with the temporal cortex
ical method used [70]. This work did not identify DHEA and EpiA level and that CSF DHEA is elevated in AD and positively correlates
oxygenated metabolites, and these results differ from several pre- with neuropathological disease stage [78]. Another report on DHEA
vious reports [71,72] that detected pregnenolone sulfate in rat derivatives in the human ventricular CSF identified 7a-HO-DHEA,
brain. 7b-HO-DHEA, 3b,7a,17b-AET, and 3b,7b,17b-AET. However, the en-
More recently, in another study, both 7a-hydroxy and 7b-hy- try site of DHEA metabolites into ventricular CSF and their role in
droxy-DHEA were generated by incubation of DHEA with rat brain this compartment are not known [79].
microsomes; 7a-hydroxy-DHEA was the major metabolite [73].
Investigators have demonstrated that 11beta-HSD1 may convert
7a-HO-DHEA to 7b-HO-DHEA in the liver and other tissues 3. Biological activities of DHEA and EpiA metabolites
[17,74,75], but whether the same mechanism for the conversion
from 7a-HO-DHEA into 7b-HO-DHEA also occurs in the brain is 7-Oxygenated steroids are widespread in mammals, birds, fish,
not yet known; future studies should focus on clearly identifying and plants [67]. The significant bioactivity of DHEA and its metab-
the enzyme(s) responsible for the conversion of 7a-HO-DHEA to olites has been noted in previous studies. This is relevant particu-
7b-HO-DHEA [59]. It should be noted that no unknown P450 cyto- larly for the 7a- and 7b-hydroxylated metabolites of DHEA and
chrome has been shown to be responsible for the direct 7beta- other 17-oxosteroids [40,80,81]. DHEA and its metabolites are
hydroxylation of DHEA [76]. To the best of our knowledge, there responsible for the above-mentioned immunoprotective and
are currently no data demonstrating the presence of 7-oxo-DHEA neuroprotective effects. However, evidence has accumulated that
in the brain (Fig. 8). 7-oxygenated DHEA derivatives may act as potent local immuno-
protective agents, which are in fact responsible for many immuno-
2.4. DHEA metabolites in cerebrospinal fluid modulatory and antiglucocorticoid effects previously ascribed to
DHEA [80]. Thus, most of the functions initially attributed to DHEA
Levels of 7a-hydroxy-DHEA, 7b-hydroxy-DHEA, and 16a-hy- from observations in rodents are now thought to be due to these
droxy-DHEA in cerebrospinal fluid (CSF) have been reported, and oxygenated metabolites.
the levels of these neuro-steroids in CSF may be helpful in the diag- As DHEA is a precursor for the metabolite EpiA, we present here
nosis of neurodegenerative diseases and in differentiating Alzhei- a single paragraph on the biological properties of DHEA and EpiA
mer’s disease (AD) from vascular dementia [77]. It is currently metabolites. A summary of recent findings on oxygenated DHEA
unknown whether CSF neuro-steroid levels are related to brain metabolites is presented in Table 1.
18 L.E. Kihel / Steroids 77 (2012) 10–26

3.1. DHEA metabolites and anticancer properties particularly for the 7-oxygenated metabolites of DHEA and other
17-oxosteroids [40,81]. A study by Pringle et al. demonstrated that
7-Oxygenated DHEA derivatives (7oxo-DHEA, 7a-HO-DHEA, 7b-HO-EpiA can protect against neuronal death in both in vivo and
and 7b-HO-DHEA), androsterone, epiandrosterone, and etiocholan- in vitro models of reperfusion ischemia-induced damage. However,
olone have anti-proliferative effects on human hepatocellular liver it is unclear how these 7-hydroxy steroids prevent cell damage
carcinoma cells (Hep G2), human epithelial colorectal adenocarci- [41].
noma cells (Caco 2), and human colon adenocarcinoma grade II The rate of aromatization of 4-adione and 7-hydroxylation of
cells (HT-29). Whether the pentose phosphate pathway (glucose- DHEA by different neuronal cell lines from fetal rat and mouse
6-phosphate dehydrogenase (G6PD), [82]) or the mevalonate path- brains was compared to that of embryonic rat hippocampal cells
way (3-hydroxy-3-methylglutaryl-CoA reductase (HMGR)), which in primary culture. The comparison of the metabolism of DHEA
are involved in cholesterol biosynthesis, or the mitogen-activated and 4-adione by neural cells of different origins shed light on the
protein kinase (MAPK)-mediated signal transduction pathway for relative importance of the aromatization and 7-hydroxylation
DNA synthesis were more responsible for the growth inhibition pathways in the brain and their roles in neuroprotection [90,91].
was investigated [29]. Because the growth inhibitory effects were Numerous studies have reported neuroprotective effects of
not correlated with the inhibition of G6PD nor HMGR activity, DHEA metabolites on the nervous system, even in aged animals,
some other mechanism for the growth inhibition may exist. How- but the results of these studies are not conclusive. To date, the
ever, these data do not exclude the possibility that the anti-prolif- in vivo effects of DHEA and its metabolites in the brain have not
erative effects of any one of the individual steroids may have been been fully elucidated, and the possibility that these oxygenated
mediated via the inhibition of G6PD and/or HMGR. These non- derivatives of DHEA might be products involved in neuroprotec-
androgenic DHEA metabolites may serve as chemopreventive or tion needs to be investigated further.
anti-proliferative therapies.
3b,17a-AED and 3b,17b-AED are neuro-steroids produced in
3.2.2. DHEA metabolites and memory
neuro-ectodermal tissue by the metabolism of DHEA. The biologi-
Studies have been carried out to assess the neuroprotective ef-
cal actions of these two epimers are distinct. 3b,17a-AED can in-
fects of DHEA and its metabolites and their ability to enhance
duce apoptosis in human lymphoma cells and autophagic cell
memory and learning in humans and animals [89,92–95].
death in malignant glioma cells, and the antiproliferative functions
7-oxo-DHEA has been shown to reverse scopolamine-induced
of this epimer are not dependent on either estrogen or androgen
memory impairment in young mice [96]. A significant and selec-
receptors [83–85]. In contrast, 3b,17b-AED enhances the immune
tive decrease in hippocampal CYP7B bioactivity was demonstrated
system with minimal anti-tumor activity [86]. More recently, one
in 24-month-old rats with impaired spatial memory performance
study indicated that autophagy in human malignant glioma cells
in the water maze but not in age-matched cognitively intact rats
and transformation of fibroblasts by treatment with the neuro-ste-
[97]. Further, treatment of cognitively impaired aged rats with
roid 3b,17a-AED proceeds through an endoplasmic reticulum
the CYP7B product 7a-hydroxypregnenolone was shown to en-
stress-dependent mechanism accompanied by partial activation
hance spatial memory retention after a 30 min delay in the radial
of the unfolded protein response. Moreover, these results implicate
arm water maze. In contrast, the parent steroid pregnenolone
PERK/eIF2a signaling as one of the pathways involved in 3b,17a-
had no significant effect at the dose tested. These authors suggest
AED-mediated autophagy and cell death [87]. Through the same
that if 7a-hydroxylated metabolites are more active than the par-
pathway, 3b,17a-AED and 3b,7a,17a-AET induce apoptosis in
ent steroid, this may explain why oral DHEA replacement in the el-
U937 lymphoma cells and autophagy in T98G glioblastoma cells.
derly has not successfully improved memory [98,99] while DHEA
These results indicate that the position of the hydroxyl group on
administration is effective at improving memory and mood in
C-17 in the a-configuration dictates the anti-tumor activity of
healthy young subjects [100], as young subjects have greater hip-
the androstene. The addition of a third hydroxyl in the a-position
pocampal CYP7B activity. Perhaps the 7a-hydroxylated metabolite
on C-7 did not increase the antitumor activity of 3a,17a-AED but
rather than the parent steroid would be a more effective therapy in
rather reduced the potency by approximately two-fold. These find-
the elderly.
ings demonstrate a strict structure–function relationship in the
anti-tumor activity for androstenediol. Moreover, 3a,17a-AED
and 3a,7a,17a-AET retain their abilities to induce autophagic cell 3.2.3. DHEA metabolites and neurodegenerative diseases
death in glial tumors, and apoptosis prevails in lymphomas, indi- Dementia, a hallmark of the aging central nervous system, has
cating that the mechanism of cell death for these two steroids is been partially attributed to a lack of circulating steroids, such as
dependent upon the origin of the tumor tissue [88]. DHEA, in several studies [101]. The pathogenesis of age-associated
disorders, such as neurodegenerative diseases, with a particular
3.2. DHEA metabolites and their effects on the brain focus on Alzheimer’s disease (AD), and their connection with DHEA
and its metabolites will be discussed here. The most frequent cause
3.2.1. DHEA metabolites and neuroprotective effects of dementia is AD (more than 60%) [102], which is a progressive
Neuroprotective effects of DHEA metabolites have been docu- neurodegenerative disorder with gradual onset and deterioration
mented both in vitro and in vivo. In the central nervous system, of cognitive functions, such as memory, language and visuo-spatial
3a,17b-AED is believed to play a neuroprotective role, but a biolog- skills [103].
ical function for 3b,17a-AED has not been identified [68]. It has Hormone replacement therapy with DHEA is a frequently dis-
been suggested that the 7-hydroxylated metabolites of DHEA cussed topic, especially in AD [1]. DHEA is a well-known inhibitor
should be considered as possible mediators of some of the neuro- of glucose 6-phosphate dehydrogenase (G6PD), and it is hypothe-
protective effects of DHEA in the brain [89]. 7a- and 7a-HO-DHEA sized that reduced G6PDH activity has a beneficial effect on age-
have neuroprotective potency mediated by antiglucocorticoid related disease development and longevity [104]. Because NADPH
activity, which once again confirmed the hypothesis that the 7- is a key cofactor in the activity of many antioxidative and reductive
hydroxylation process may explain the neuroprotection offered enzymes [105], its depletion may result in an impairment of fuel
by neuro-steroids [56]. utilization and consequently the onset of the disease. In addition
Recent reports indicate that DHEA metabolites are the local to its other roles, NADPH is a co-factor for the enzyme CYP7B1,
agents responsible for the neuroprotective effects. This is relevant which is abundant in brain tissues and responsible for
L.E. Kihel / Steroids 77 (2012) 10–26 19

7a-hydroxylation of DHEA [106]. The expression of this enzyme in (RA), has also been investigated [115]. Within 1 day of culture with
post-mortem brain tissues is decreased in AD [107]. radiolabeled DHEA, monocyte-derived macrophages converted
The metabolism of DHEA was recently analyzed in the aging DHEA to D5-derivatives, such as 16a-hydroxy-DHEA, 3b,17b-
brain from Alzheimer patients and non-demented controls. The AED, and 3b, 16a, 17b-AET. The conversion of DHEA leads to in-
conversion of DHEA to 3b,17b-AED and to 7a-OH-DHEA occurs in creased downstream effector hormones in target macrophages,
the frontal cortex, hippocampus, amygdala, cerebellum and stria- which may be an important factor for local immunomodulation.
tum of both Alzheimer’s patients and controls. The formation of The conversion of DHEA to downstream effector hormones and
these metabolites within distinct brain regions is negatively corre- the regulation of key enzymes in macrophages may explain some
lated with the density of b-amyloid deposits [108]. of the effects of DHEA therapy in chronic inflammatory diseases
Epiandrosterone is also converted to 7b-OH-EpiA in numerous or arteriosclerosis. A study on DHEA metabolism in arthritis
tissues. Indeed, DHEA and epiandrosterone (EpiA), which is derived showed that the severity of arthritis correlates with increased lev-
from DHEA, are ineffective in protecting organotypic hippocampal els of the P450 enzyme CYP7B, which converts DHEA into7a-hy-
slice cultures against hypoxia/reperfusion-induced neuronal cell droxy-DHEA [116]. Increased CYP7B activity leads to higher
death, whereas their corresponding 7-hydroxy derivatives had levels of the DHEA metabolite hydroxy-DHEA in synovial fluid,
powerful neuroprotective effects [40]. Therefore, 7a-hydroxy which may contribute to the maintenance of the chronic inflam-
DHEA and 7b-hydroxy EpiA may be endogenous neuroprotective mation observed in rheumatoid arthritis patients [117,118].
agents, and 7-hydroxylation of DHEA and EpiA may be impaired 3b,7b,17b-AET was described previously to be a more highly
in neurodegenerative conditions, such as AD. oxygenated natural DHEA metabolite found in humans [119] that
Other findings indicate that 7b-HO-DHEA has powerful cytopro- provides anti-inflammatory benefits in animal models of autoim-
tective effects, suggesting that this neuro-steroid may have thera- munity and trauma [120–125].
peutic potential in various neurodegenerative conditions, such as In rodents, DHEA is metabolized to produce (among other com-
AD, and 7b-hydroxy steroids may constitute a novel class of endog- pounds) 3b,7b,17b-AET, which has potent anti-inflammatory activ-
enous neuroprotective agents [41]. It was also reported that ity. Its functions in the body may include tissue-specific
3b,17b-AED may regulate the cerebral levels of DHEA and may con- modulation of glucocorticoid action, immune function, and control
tribute to the control of DHEA activity in the aging brain and in Alz- of acute and chronic inflammation [122,123]. The anti-inflamma-
heimer’s disease [61,109]. tory activity of two tetrols (androst-5-ene-3b,7b,16a,17b-tetrol
DHEA and DHEAS have been widely studied in neurodegenera- (I) and androst-5-ene-3a,7b,16a,17b-tetrol (III, Fig. 4) was demon-
tive diseases, but the true role of oxygenated DHEA metabolites in strated in rodent models of acute and chronic inflammation. Anti-
these diseases is still not clear. Several reports on DHEA metabo- inflammatory properties were observed with exogenous adminis-
lites have suggested their neuroprotective effects, but these have tration of these compounds in rodent disease models of multiple
not been proven. Further research is necessary to clarify this topic. sclerosis, lung injury, chronic prostatitis, and colitis [39].

3.3. DHEA metabolite effects on inflammatory and immune processes 3.4. DHEA metabolites and osteoporosis

The antioxidant effects of DHEA and 7a-hydroxy-DHEA against In rodents, DHEA opposes certain activities of endogenous glu-
oxidative stress induced by colitis were investigated in vivo in rats. cocorticoids, such as osteoporosis [126,127], and improves osteo-
DHEA and 7a-hydroxy-DHEA exerted a significant anti-oxidant ef- blast growth and bone tissue morphometry [128]. As the
fect against oxidative stress induced colitis in rats, reducing oxida- literature grew, DHEA became widely used as an anti-aging and
tive damage to proteins and lipids. These findings demonstrate anti-stress dietary supplement and was considered a promising
that DHEA does not act alone and that its 7-hydroxylated metabo- agent to treat osteoporosis. Despite promising results in rodents,
lites may be of importance for its protective effects [110,111]. DHEA provided only modest improvement in bone mineral density
Strong anti-oxidant and anti-inflammatory properties of 7b-hy- (BMD) in patients treated with GC and elicited unwanted andro-
droxy-EpiA have been reported in vivo in rats. Although all dose genic side effects [129–131].
levels of 7b-hydroxy-EpiA reduced prostaglandin E2 (PGE2) pro- The discovery of the receptor activator of nuclear factor-jB li-
duction, only the lowest dose (0.01 mg/kg) completely prevented gand (RANKL)/RANK/osteoprotegerin system and its role in the
colitis damage and tissue inflammation. 7b-Hydroxy-EpiA pre- regulation of bone resorption exemplifies how both serendipity
treatment prevents the occurrence of sodium dextran sulfate-in- and a logic-based approach can identify factors that regulate cell
duced colitis through a shift from PGE2 to PGD2 production [112]. function.
A more recent study demonstrated an important role for 7b-hy- The study examined the particular effects of a and b androst-5-
droxy-EpiA in the regulation of cytoprotective prostaglandins from ene steroid epimers (3b,17b-AED, 3b,7b,17b-AET, 3b,17a-AED, and
human mononuclear cells. An elevation in the circulating levels of 3b,7a,17b-AET) on immortalized human fetal osteoblasts in cul-
these prostanoids may be important in the neuroprotective actions ture and examined the ability of a/b androstenediol metabolites
of 7b-hydroxy-EpiA [113]. to signal effects by regulating PPAR in osteoblasts. The results
The effects of 7a-hydroxy-DHEA, 7b-hydroxy-DHEA and 7b-hy- showed that 3b,17a-AED opposes 3b,17b-AED by elevating bone
droxy-EpiA on prostaglandin (PG) production and related enzyme resorption in osteoblast cells. The increase in RANKL/OPG is mod-
gene expression were tested. Human peripheral blood monocytes ulated by the activation of PPAR-c that in turn caused apoptosis of
were activated by incubation with the inflammatory cytokine FOB-9 cells [132].
TNF-a in the presence and absence of these hydroxysteroids. The The potential role of 3b,7b,17b-AET in bone formation is high-
results suggest that 7b-hydroxy-EpiA produced locally at the site lighted by these in vitro findings related to 3b,7b,17b-AET-induced
of inflammation may trigger a shift in the PG pathway, favoring in- OPG expression in MG-63 osteoblasts treated with dexametha-
creased production of 15d-PGJ2 at the expense of PGE2. The pro- sone, a synthetic glucocorticoid. OPG is a soluble decoy RANK
tective effects of 15d-PGJ2 on neighboring cells could contribute receptor that is secreted by osteoblasts and binds to the surface
to the resolution of inflammation and tissue repair, thus facilitating of RANK ligand (RANKL) on osteoblasts. This decreases the pool
the immune response [114]. of RANKL available for RANK receptor binding, decreasing the acti-
The conversion of DHEA to downstream steroid hormones in vation of the RANK receptor on pre-osteoclasts and ultimately
target macrophages, as major effector cells in rheumatoid arthritis osteoclast commitment and differentiation [133].
20 L.E. Kihel / Steroids 77 (2012) 10–26

Recent findings showed that 3b,7b,17b-AET preserves bone androst-5ene-3b,7a,17b-triol) on the expression, accumulation,
mineral content, but not mineral density, from thermal trauma-in- and catalytic activity of key lipid-metabolizing enzymes in 3T3-
duced bone loss, and it limits cortical cancellous bone loss and pre- L1 cells as the experimental model was examined [147]. This work
serves the endochondral growth rate but not cortical bone loss showed that 7-oxo-DHEA has an opposite effect on adipocyte dif-
[134]. ferentiation and thermogenesis. These disparate effects suggest
that DHEA and 7-oxo-DHEA may be targeting different aspects of
3.5. DHEA metabolites and thermogenesis the differentiation program.
Another study identified and quantitated DHEA metabolites
Thyroid hormones are the main hormonal regulators of in adipose cells (3T3-L1) by LC–MS analysis. These results
thermogenesis. demonstrated that adipocytes rapidly converted DHEA to and-
Feeding DHEA to rats enhances heat production and decreases rost-5-ene-3b,17b-diol. 7a-Hydroxy-DHEA was interconverted
the efficiency of food utilization. The effects of DHEA were mea- with 7-oxo-DHEA and 7b-hydroxy-DHEA and the corresponding
sured by the production of liver mitochondrial sn-glycerol-3-phos- 17b reduced products (Fig. 9). DHEA and its derivatives were
phate dehydrogenase and cytosolic malic enzyme, two enzymes detected only in the culture medium, suggesting that DHEA is
that had been demonstrated to be induced by the classic thermo- metabolized via peripheral or integral cell membrane enzymes.
genic agent thyroid hormone [135,136]. Alternatively, there may be efficient mechanisms at play for the
Derivatives of DHEA were investigated for their ergogenic prop- secretion of the steroids to the aqueous media rather than reten-
erties, and 7-oxo-DHEA in particular was shown to be 4-fold more tion in the lipid-rich cell. Interestingly, the authors identified
thermogenic than DHEA [137]. Both 7-hydroxy-DHEA epimers and 3b,17b-AED as the major metabolite of DHEA [148].
7-oxo-DHEA are more thermogenic than DHEA itself. The thermo-
genic properties of DHEA and 7-oxo-DHEA are likely to be due to 3.7. DHEA metabolites and the skin
two interrelated actions: increased levels of enzymes and proteins
that shuttle substrate and electrons in and out of the mitochondria Similar to the classical steroidogenic organs, such as gonads and
and an increased proton leak across the mitochondrial inner mem- adrenal glands, the skin and its appendages, including hair follicles,
brane [138]. There is greater experimental evidence for increased sebaceous glands, and eccrine/apocrine glands, are armed with all
shuttling by DHEA and 7-oxo-DHEA than for induction of a proton of the necessary enzymes required for androgen synthesis and
leak. The levels of malic enzyme, which converts pyruvate to ma- metabolism [149]. However, after cessation of estrogen secretion
late, can be increased more than 5-fold by 7-oxo-DHEA [139]. In by the ovaries at menopause, practically all sex steroids acting in
further experiments, mitochondria from rats treated with 7-oxo- the skin are synthesized locally from DHEA [150]. Immunohisto-
DHEA appear to have an increased proton leak across the inner chemical studies have demonstrated the presence of both CYP7B1
mitochondrial membrane, similar to thyroid hormone [140]. Fur- and 11beta-HSD1 in human skin and other tissues [17].
thermore, plasma levels of DHEA and its metabolites, such as Some effects of DHEA on human skin, including collagen gene
3b,17a-AED, may be altered in thyroid disorders [141]. expression and protein synthesis, have already been described
Previous studies have attempted to establish a relationship be- [151–155]. For example, recent findings strongly suggest the pos-
tween thyroid laboratory parameters, including major thyroid sibility that DHEA exerts anti-aging effects in the skin through
auto-antibodies and DHEA and its 7-hydroxylated metabolites, stimulation of collagen biosynthesis and improved structural orga-
which are believed to act as immunoprotective agents [80,142]. nization of the dermis while modulating keratinocyte metabolism
Recently, it was demonstrated that administration of 7-oxo-DHEA, [154].
one of the candidates of steroid replacement therapy, to healthy The major product of DHEA metabolism in EpiSkin is DHEAS
male volunteers temporarily influenced the actual levels of thyroid (88%), while the other metabolites are 7a-HO-DHEA (8.2%), 4-adi-
hormones [143]. one (1.3%), 3b,17b-AED (1.3%), dihydrotestosterone (DHT) (1.4%)
Other data clearly demonstrates a negative relationship be- and androsterone (ADT) (2.3%) [153]. However, the role of oxygen-
tween the levels of the immunoprotective steroids (7a-HO- and ated DHEA metabolites in the prevention of skin aging has not been
7b-HO-DHEA) and the extent of the autoimmune process, at least clearly established.
in hypothyroidism [144].
3.8. DHEA metabolites and the cardiovascular system
3.6. DHEA metabolites and adipogenesis
Previous work has shown that left ventricular performance, car-
Adipocytes have been intensively studied in recent years, and diac output, and organ blood flow in the liver, small intestine, and
the mouse embryo-derived 3T3-L1 cell line has often been used kidney decreased significantly after trauma-induced hemorrhage
as a model system for the study of preadipocyte differentiation, [156–158].
maturation, and cellular processes [145]. One study indicated that 3b,17b-AED administration after trau-
Adipose tissue contains significant quantities of DHEA (10- ma-induced hemorrhage improves cardiovascular function and or-
times greater than in the circulation) and possesses the appropri- gan blood flow and decreases cytokine production in male animals.
ate enzymatic machinery for the metabolism of DHEA. Thus, inves- This improvement of cardiovascular function may be associated
tigators have examined a role for DHEA in adipose tissue biology, with attenuation in IL-6 levels and an increase in plasma nitrite/ni-
and despite the low circulating levels of DHEA/DHEAS in rodents, trate levels. Because 3b,17b-AED produced the above-mentioned
many have used murine cell lines or in vivo rodent models as their effect, 3b,17b-AED appears to be a useful adjuvant for restoring de-
experimental systems. DHEA has been reported to inhibit 3T3-L1 pressed cardiovascular function and attenuating cytokine produc-
adipogenesis and proliferation [146]. tion after trauma-induced hemorrhage [159,160]. To the best of
The anti-adipogenic effect of DHEA on human omental pre-adi- our knowledge, only the effect of 3b,17b-AED has been studied.
pocytes in vitro is in agreement with the significant decrease in
abdominal fat in elderly subjects treated with DHEA for 6 months 3.9. DHEA metabolites and estrogenic receptors
[4].
The effect of DHEA and 7-oxygenated metabolites (7-oxo-DHEA, Estrogens exert a number of biological effects throughout the
7a-OH-DHEA, 7b-OH-DHEA, androst-5ene-3b, 7b,17b-triol and body, most of which are mediated via the estrogen receptors ERa
L.E. Kihel / Steroids 77 (2012) 10–26 21

O
OH O

HO HO
HO
3β,17β−AED Testosterone
DHEA

O
OH

4-Adione
HO OH
HO OH

7α-HO-DHEA
3β,7α,17α−AET

OH
O O

HO OH
HO O HO OH
3β,7β,17β−AET
7-oxo-DHEA 7β-HO-DHEA
Fig. 9. Proposed metabolic route for metabolism of DHEA in differentiating 3T3-L1 adipocytes. Modified scheme from Marwah et al. [148].

and b. Estrogens play important functions in the female as well as 173]. Other studies have suggested that when there is low metab-
the male reproductive system and in several non-reproductive tis- olism of DHEA or if 4-adione and 3b,17b-AED are produced, andro-
sues. For instance, it has been proposed that ERa might stimulate genic effects are observed, while other metabolites (DHEA-S,
proliferation whereas ERb inhibits proliferation [161,162]. DHEA 7a-HO-DHEA, 7b-HO-DHEA, and 7-oxo-DHEA) essentially lack
and its metabolites have been evaluated as ligand activators of androgenic activities [174].
ERa and ERb [163,164]. In some studies, it has been proposed that
activation of ERb by 5a-androstane-3b,17b-diol (3b,17b-AAD) may
3.11. DHEA metabolites and the human prostate
be important for suppression of prostate growth [165].
Because 3b,17b-AAD is metabolized by CYP7B1, it has been sug-
The prostate is an androgen-dependent organ, the growth,
gested that this enzyme might decrease ERb action via control of
maintenance and secretory functions of which are stimulated by
3b,17b-AAD levels. In contrast, other investigators have reported
the continuous presence of androgens and growth factors. In hu-
that a steroid metabolite formed by CYP7B1, 7a-hydroxy-DHEA,
mans, the adrenal glands secrete a large amount of the precursor
activates ERb and that CYP7B1 catalysis therefore should increase
steroid DHEA, which can be converted into androst-4-ene-3,17-
ERb action [166]. Furthermore, a cholesterol derivative metabo-
dione or androst-5-ene-3b,17b-diol and then testosterone (T) and
lized by CYP7B1, 27-hydroxycholesterol, was recently found to ex-
further metabolized into the potent androgen 5a-dihydrotestos-
ert pro-estrogenic actions in some cells but suppress ER activation
terone in prostatic tissues [175].
in others [167,168]. Recent data indicate that low concentrations of
Various steroidogenic enzymes in the prostate are involved in
3b,17b-AED can trigger ER-mediated responses equally well for
the biosynthesis and metabolism of steroid precursors, such as
both ERa and b and that CYP7B1-mediated conversion of 3b,17b-
DHEA [176], and steroidogenesis could differ in different species,
AED into a 7a-hydroxy-metabolite will result in a loss of activity
such as human, dog, rat and mouse prostate tissues, as well as pri-
[169].
mary cells and prostatic cancer cell lines [177]. The in vitro DHEA
metabolites formed from human prostate homogenates were
3.10. DHEA metabolites and androgenic receptors identified by using LC–MS and GC–MS techniques. Androst-
5-ene-3b,17b-diol was the main DHEA metabolite identified.
A ligand sensitive androgen receptor (AR) is involved in the reg- 7a-hydroxy-DHEA, 7a-hydroxy-DHEA, 7-oxo-DHEA, androst-4-
ulation of prostate growth, muscle and bone mass, and spermato- ene-3,17-dione, testosterone, and 5a-dihydrotestosterone were
genesis in males. AR agonists are therapeutically useful in the identified as well [178].
treatment of osteoporosis, cachexia, contraception, and androgen Androgens are involved in many diseases, especially benign
deficiency [168,170], and antagonists are used for the treatment prostatic hyperplasia and prostate cancer. However, few reports
of prostate cancer. It has been shown that 3b,17b-AED is a strong have investigated the levels of adrenal androgens in prostate can-
activator of AR target genes, but DHEA and 7-oxo-DHEA cannot cer tissues after androgen deprivation [179].
activate AR target genes in the absence or presence of the co-acti- In recent studies, analyses of steroids were performed using LC–
vator ARA70 in DU145 cells co-transfected with a reporter gene MS in castration-resistant prostate cancer tissues after androgen
and expression plasmid (wtAR, mtAR877, or mtAR708) [171– deprivation therapy. The levels of DHEA, androst-4-ene-3,
22 L.E. Kihel / Steroids 77 (2012) 10–26

17-dione, androst-5-ene-3b,17b-diol, testosterone and dihydrotes- species with as yet undescribed chemical and biological activities.
tosterone in prostate cancer were detected by using LC–MS/MS for Once considered to be physiologically inactive, these species repre-
a precise simultaneous quantitative analysis. They found that the sent a novel pharmacological target for designing new DHEA ana-
levels of DHEA were high in prostate cancer tissues, irrespective logs with potent biological activity that might be devoid of
of androgen deprivation therapy. These authors suggested that unwanted estrogenic or androgenic side effects. These data should
DHEA plays a significant role in the synthesis of T and DHT in pros- be explored for the development of a specific synthetic derivative
tate cancer tissues after androgen deprivation therapy [180]. that is less labile to metabolism and may at last deliver the benefits
It should be noted that androst-5-ene-3b,17b-diol is found at a of oxygenated DHEA metabolites observed in animals and humans.
high concentration in prostate cancer tissue, even after androgen Thus, synthetic derivatives of oxygenated DHEA metabolites could
deprivation therapy. Indeed, its androgen activity is not inhibited be used as a possible platform to derive novel pharmaceutical
by the antiandrogens currently used to treat prostate cancer pa- products.
tients [173].
DHEA can directly activate the androgen receptor (AR) or estro-
gen receptor beta (ERb) in the prostatic epithelium or the AR or References
estrogen receptor alpha (ERa) in the prostatic stroma. Indirect ef-
fects are conferred by DHEA metabolism of androgenic ligands [1] Cameron DR, Braunstein GD. The use of dehydroepiandrosterone therapy in
clinical practice. Treat Endocrinol 2005;4(2):95–114.
(including androstenedione, testosterone and dihydrotestosterone) [2] Bovenberg SA, van Uum SH, Hermus AR. Dehydroepiandrosterone
or estrogenic ligands (including 7-OH-DHEA, 3b-adiol, or 17b- administration in humans: evidence based? Neth J Med 2005;63(8):300–4.
estradiol) [181]. Alternatively, DHEA metabolites may act on ERb [3] Oberbeck R, Kobbe P. Dehydroepiandrosterone (DHEA): a steroid with
multiple effects. Is there any possible option in the treatment of critical
in the prostate, potentially antagonizing androgenic effects on illness? Curr Med Chem 2010;17(11):1039–47.
prostate cancer growth, such as DHEA metabolism into 7a-hydro- [4] Villareal DT, Holloszy JO. Effect of DHEA on abdominal fat and insulin action
xy-DHEA (7HD), a known ligand for ERb [182]. The complexity of in elderly women and men: a randomized controlled trial. J Am Med Assoc
2004;292(18):2243–8.
the balance between androgenic and estrogenic effects, whether [5] Talaei A, Amini M, Siavash M, Zare M. The effect of dehydroepiandrosterone
as direct ligands or DHEA metabolites on the prostate, is matched on insulin resistance in patients with impaired glucose tolerance. Hormones
by the complexity of estrogen action through ERa and ERb. The 2010;9(4):326–31.
[6] Tok EC, Ertunc D, Oz U, Camdeviren H, Ozdemir G, Dilek S. The effect of
intratissular balance of androgen and estrogen levels is most circulating androgens on bone mineral density in postmenopausal women.
important for prostatic development and differentiation [183– Maturitas 2004;48(3):235–42.
185]. Many questions remain to be clarified on the regulation of [7] Juhasz-Vedres G, Rozsa E, Rakos G, et al. Dehydroepiandrosterone sulfate is
neuroprotective when administered either before or after injury in a focal
DHEA metabolism in the prostate. Oxidative metabolism of DHEA
cortical cold lesion model. Endocrinology 2006;147:683–6.
has not been fully investigated either in primary cells or prostatic [8] Wang L, Hao Q, Wang YD, Wang WJ, Li DJ. Protective effects of
cancer cell lines [186–189]. This unresolved question remains: dehydroepiandrosterone on atherosclerosis in ovariectomized rabbits via
what biochemical mechanism promotes the androgenic and estro- alleviating inflammatory injury in endothelial cells. Atherosclerosis
2011;214(1):47–57.
genic metabolism of DHEA? [9] Celec P, Starka L. Dehydroepiandrosterone - is the fountain of youth drying
out? Physiol Res 2003;52:397–407.
[10] Zheng P. Neuroactive steroid regulation of neurotransmitter release in the
CNS: action, mechanism and possible significance. Prog Neurobiol
4. Conclusion 2009;89(2):134–52.
[11] Labrie F, Luu-the V, Labrie C, Simard J. DHEA and its transformation into
In recent years, significant interest has arisen from the hypoth- androgens and estrogens in peripheral target tissues: Intracrinology. Front
Neuroendocrinol 2001;22:185–212.
eses that declining DHEA concentrations in adults may serve as an [12] Labrie F. DHEA, important source of sex steroids in men and even more in
indicator of a number of pathological conditions associated with women. Prog Brain Res 2010;182:97–148.
aging, many of which are correlated with changes in lipid metabo- [13] Fitzpatrick JL, Ripp SL, Smith NB, Pierce WM, Prough RA. Metabolism of DHEA
by cytochromes P450 in rat and human liver microsomal fractions. Arch
lism. However, the precise role of DHEA and its metabolites re- Biochem Biophys 2001;389:278–87.
mains uncertain in spite of many epidemiological and biological [14] Marwah A, Marwah P, Lardy H, Ergosteroids VI. Metabolism of
studies. Many of the functions initially attributed to DHEA from dehydroepiandrosterone by rat liver in vitro: a liquid chromatographic-
mass spectrometric study. J Chromatogr B Analyt Technol Biomed Life Sci
observations in humans and animals are now thought to be prop-
2002;767(2):285–99.
erties of its oxygenated metabolites. However, the genomic effects [15] Chalbot S, Morfin R. Human liver S9 fractions: metabolism of
of DHEA and its major metabolites remain controversial because dehydroepiandrosterone, epiandrosterone, and related 7-hydroxylated
the metabolism of DHEA varies in different cells and tissues. Recent derivatives. Drug Metab Dispos 2005;33:563–9.
[16] Kim SB, Chalbot S, Pompon D, Jo DH, Morfin R. The human cytochrome
studies have described DHEA metabolism in different tissues, such P4507B1: catalytic activity studies. J Steroid Biochem Mol Biol
as in rodent and human livers, and identification of an extensive 2004;92:383–9.
number of oxygenated DHEA metabolites has been achieved. How- [17] Hennebert O, Chalbot S, Alran S, Morfin R. Dehydroepiandrosterone 7alpha-
hydroxylation in human tissues: possible interference with type 1 11beta-
ever, the major metabolic pathway for DHEA in extra-hepatic tis- hydroxysteroid dehydrogenase-mediated processes. J Steroid Biochem Mol
sues has not fully been elucidated. Indeed, the study of the Biol 2007;104:326–33.
biochemical pathway of DHEA metabolites in the brain is an attrac- [18] Trap C, Nato F, Chalbot S, Kim SB, Lafaye P, Morfin R. Immunohistochemical
detection of the human cytochrome P4507B1: production of a monoclonal
tive alternative for exploring the pathways involved in the patho- antibody after cDNA immunization. J Neuroimmunol 2005;159:41–7.
genesis of neurodegenerative diseases. [19] Muller C, Pompon D, Urban P, Morfin R. Inter-conversion of 7alpha- and
Further characterization of the physiological and molecular 7beta-hydroxy-dehydroepiandrosterone by the human 11beta-
hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol
mechanisms of DHEA and its metabolites would help to clarify 2006;99:215–22.
their potential roles in cellular metabolism. Moreover, elucidating [20] Morfin R, Courchay G. Pregnenolone and dehydroepiandrosterone as
the molecular mechanisms of DHEA and its oxygenated metabo- precursors of native 7-hydroxylated metabolites which increase the
immune response in mice. J Steroid Biochem Mol Biol 1994;50(1–2):91–100.
lites in preventing the initiation of aging of the brain and periphe-
[21] Hennebert O, Montes M, Favre-Reguillon A, Chermette H, Ferroud C, Morfin R.
ral tissues would offer immense opportunities for the development Epimerase activity of the human 11beta-hydroxysteroid dehydrogenase type
of future treatments. 1 on 7-hydroxylated C19-steroids. J Steroid Biochem Mol Biol 2009;114(1–
For instance, the diversity of highly oxygenated steroids identi- 2):57–63.
[22] Lootens L, Van Eenoo P, Meuleman P, Leroux-Roels G, Delbeke FT. The uPA(+/
fied in the human liver raises the possibility of naturally occurring +)-SCID mouse with humanized liver as a model for in vivo metabolism of 4-
diols, triols and tetrols and perhaps even more highly oxygenated androstene-3, 17-dione. Drug Metab Dispos 2009;37(12):2367–74.
L.E. Kihel / Steroids 77 (2012) 10–26 23

[23] Katoh M, Tateno C, Yoshizato K, Yokoi T. Chimeric mice with humanized liver. [49] Do Rego JL, Tremblay Y, Luu-The V, Repetto E, Castel H, Vallarino M, et al.
Toxicology 2008;246(1):9–17. Immunohistochemical localization and biological activity of the
[24] Kerkhof EG, Graaf IA, Jager MH, Groothuis GM. Induction of phase I and II drug steroidogenic enzyme cytochrome P450 17alpha-hydroxylase/C17, 20-lyase
metabolism in rat small intestine and colon in vitro. Drug Metab Dispos (P450C17) in the frog brain and pituitary. J Neurochem 2007;100(1):251–68.
2007;35:898–907. [50] Tsutsui K, Matsunaga M, Miyabara H, Ukena K. Neurosteroid biosynthesis in
[25] Kerkhof EG, Ungell AB, Sjoberg AK, Jager MH, Hilgendorf C, Graaf IAM, the quail brain: a review. J Exp Zool A Comp Exp Biol 2006;305(9):733–42.
Groothuis GMM. Innovative methods to study human intestinal drug [51] Schumacher M, Akwa Y, Guennoun R, Robert F, Labombarda F, Desarnaud F,
metabolism in vitro: precision-cut slices compared with Ussing Chamber et al. Steroid synthesis and metabolism in the nervous system: trophic and
preparations. Drug Metab Dispos 2006;34:1893–902. protective effects. J Neurocytol 2000;29(5–6):307–26.
[26] Wang S, Rijk JC, Riethoff-Poortman JH, Van Kuijk S, Peijnenburg AA, Bovee TF. [52] Mellon SH, Griffin LD. Neurosteroids: biochemistry and clinical significance.
Bovine liver slices combined with an androgen transcriptional activation Trends Endocrinol Metab 2002;13:35–43.
assay: an in-vitro model to study the metabolism and bioactivity of steroids. [53] Hojo Y, Hattori TA, Enami T, Furukawa A, Suzuki K, Ishii HT, Mukai H,
Anal Bioanal Chem 2010;397(2):631–41. Morrison JH, Janssen WG, Kominami S, Harada N, Kimoto T, Kawato S. Adult
[27] Lardy H, Marwah A, Marwah P. Transformations of DHEA and its metabolites male rat hippocampus synthesizes estradiol from pregnenolone by
by rat liver. Lipids 2002;37(12):1187–91. cytochromes P45017a and P450 aromatase localized in neurons. Proc. Natl
[28] Stevens JC, Hines RN, Gu C, Koukouritaki SB, Manro JR, Tandler PJ, Zaya MJ. Acad Sci USA 2004;101:865–70.
Developmental expression of the major human hepatic CYP3A enzymes. J [54] Cascio C, Brown RC, Hales DB, Papadopoulos V. Pathways of
Pharmacol Exp Ther 2003;307:573–82. dehydroepiandrosterone formation in rat brain glia. J Steroid Biochem Mol
[29] Yoshida S, Honda A, Matsuzaki Y, Fukushima S, Tanaka N, Takagiwa A, et al. Biol 2001;75:177–86.
Anti-proliferative action of endogenous dehydroepiandrosterone metabolites [55] Liu Y, Pocivavsek A, Papadopoulos V. Dehydroepiandrosterone formation is
on human cancer cell lines. Steroids 2003;68(1):73–83. independent of cytochrome P450 17alpha-hydroxylase/17, 20 lyase activity
[30] Robinzon B, Miller KK, Prough RA. Biosynthesis of [3H]7 alpha-hydroxy-, 7 in the mouse brain. J Steroid Biochem Mol Biol 2009;115(3–5):86–90.
beta-hydroxy-, and 7-oxo-dehydroepiandrosterone using pig liver [56] Morfin R, Starka L. Neurosteroid 7-hydroxylation products in the brain. Int
microsomal fractions. Anal Biochem 2004;333(1):128–35. Rev Neurobiol 2001;46:79–95.
[31] Baker ME. Co-evolution of steroidogenic, steroid-inactivating enzymes, [57] Steckelbroeck S, Watzka M, Lütjohann D, Makiola P, Nassen A, Hans VHJ, et al.
adrenal, sex steroid receptors. Mol cell Endocrinol 2004;215(1–2):55–62. Characterization of the dehydroepiandrosterone (DHEA) metabolism via
[32] Nashev LG, Chandsawangbhuwana C, Balazs Z, Atanasov AG, Dick B, Frey FJ, oxysterol 7a-hydroxylase and 17-ketosteroid reductase activity in the human
Baker ME, Odermatt A. Hexose-6-phosphate dehydrogenase modulates brain. J Neurochem 2002;83:713–26.
11beta-hydroxysteroid dehydrogenase type 1-dependent metabolism of 7- [58] Stoffel-Wagner B. Neurosteroid metabolism in the human brain. Eur J
keto- and 7beta-hydroxy-neurosteroids. PLoS One 2007;2(6):e561. Endocrinol 2001;145:669–79.
[33] Rijk JCW, Bovee TFH, Groot MJ, Peijnenburg AACM, Nielen MWF. Evidence of [59] Li A, Bigelow JC. The 7-hydroxylation of dehydroepiandrosterone in rat brain.
the indirect hormonal activity of prohormones using liver S9 metabolic Steroids 2010;75(6):404–10.
bioactivation and an androgen bioassay. Anal Bioanal Chem [60] Rose KA, Stapleton G, Dott K, Kieny MP, Best R, Schwarz M, et al. Cyp7b, a
2008;392(3):417–25. novel brain cytochrome P450, catalyzes the synthesis of neurosteroids 7a-
[34] Robinzon B, Michael KK, Ripp SL, Stephen J, Winters SJ, Prough RA. hydroxy dehydroepiandrosterone and 7a-hydroxy pregnenolone. Proc Natl
Glucocorticoids inhibit interconversion of 7-hydroxy- and 7-oxo- Acad Sci USA 1997;94:4925–30.
metabolites of dehydroepiandrosterone (DHEA): a role for 11b- [61] Weill-Engerer S, David JP, Sazdovitch V, Liere P, Schumacher M, Delacourte A,
hydroxysteroid dehydrogenases? Arch Biochem Biophys 2003;412(2):251–8. et al. In vitro metabolism of dehydroepiandrosterone (DHEA) to 7alpha-
[35] Robinzon B, Prough RA. Interactions between dehydroepiandrosterone and hydroxy-DHEA and Delta5-androstene-3beta, 17beta-diol in specific regions
glucocorticoid metabolism in pig kidney: nuclear and microsomal 11beta- of the aging brain from Alzheimer’s and non-demented patients. Brain Res
hydroxysteroid dehydrogenases. Arch Biochem Biophys 2005;442(1):33–40. 2003;969(1–2):117–25.
[36] Miller KK, Cai J, Ripp SL, Pierce Jr WM, Rushmore TH, Prough RA. Stereo- and [62] Brown RC, Cascio C, Papadopoulos V. Neurosteroids: oxidative stress-
regioselectivity account for the diversity of dehydroepiandrosterone (DHEA) mediated dehydroepiandrosterone formation in Alzheimer’s disease
metabolites produced by liver microsomal cytochromes P450. Drug Metab pathology. Neurobiol Aging 2000;21:S238.
Dispos 2004;32(3):305–13. [63] Yang HY, Lee QP, Rettie AE, Juchau MR. Functional cytochrome P4503A
[37] Bellemare V, Faucher F, Breton R, Luu-the V. Characterization of 17alpha- isoforms in human embryonic tissues: expression during organogenesis. Mol
hydroxysteroid dehydrogenase activity (17alpha-HSD) and its involvement in Pharmacol 1994;46:922–8.
the biosynthesis of epitestosterone. BMC Biochem 2005;6:12. [64] Steckelbroeck S, Watzka M, Reissinger A, Wegener-Toper P, Bidlingmaier F,
[38] Odermatt A, Nashev LG. The glucocorticoid-activating enzyme 11beta- Bliesener N, Hans VH, Clusmann H, Ludwig M, Siekmann L, Klingmüller D.
hydroxysteroid dehydrogenase type 1 has broad substrate specificity: Characterisation of estrogenic 17beta-hydroxysteroid dehydrogenase
Physiological and toxicological considerations. J Steroid Biochem Mol Biol (17beta-HSD) activity in the human brain. J Steroid Biochem Mol Biol
2010;119(1–2):1–13. 2003;86(1):79–92.
[39] Ahlem CN, Page TM, Auci DL, Kennedy MR, Mangano K, Nicoletti F, Ge Y, [65] Mindnich R, Möller G, Adamski J. The role of 17 beta-hydroxysteroid
Huang Y, White SK, Villegas S, Conrad D, Wang A, Reading CL, Frincke JM. dehydrogenases. Mol Cell Endocrinol 2004;218(1–2):7–20.
Novel components of the human metabolome: the identification, [66] Mindnich R, Deluca D, Adamski J. Identification and characterization of 17
characterization and anti-inflammatory activity of two 5-androstene beta-hydroxysteroid dehydrogenases in the zebrafish, Danio rerio. Mol Cell
tetrols. Steroids 2011;76(1–2):145–55. Endocrinol 2004;215(1–2):19–30.
[40] Pringle A, Schmidt W, Deans J, Wulfert E, Reymann K, Sundstrom L. 7- [67] Lathe R. Steroid and sterol 7-hydroxylation: ancient pathways. Steroids
Hydroxylated epiandrosterone (7-OH-EPIA) reduces ischemia-induced 2002;67:967–77.
neuronal damage both in vivo and in vitro. Eur J Neurosci 2003;18:117–24. [68] Zwain IH, Yen SS. Neurosteroidogenesis in astrocytes, oligodendrocytes, and
[41] Dudas B, Hanin I, Rose M, Wulfert E. Protection against inflammatory neurons of cerebral cortex of rat brain. Endocrinology 1999;140:3843–52.
neurodegeneration and glial cell death by 7beta-hydroxy epiandrosterone, a [69] Gottfried-Blackmore A, Sierra A, Jellinck PH, McEwen BS, Bulloch K. Brain
novel neurosteroid. Neurobiol Dis 2004;15:262–8. microglia express steroid-converting enzymes in the mouse. J Steroid
[42] Hennebert O, Pernelle C, Ferroud C, Morfin R. 7Alpha- and 7beta-hydroxy- Biochem Mol Biol 2008;109(1–2):96–107.
epiandrosterone as substrates and inhibitors for the human 11beta- [70] Ebner MJ, Corol DI, Havlíková H, Honour JW, Fry JP. Identification of
hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol neuroactive steroids and their precursors and metabolites in adult male rat
2007;105:159–65. brain. Endocrinology 2006;147(1):179–90.
[43] Hennebert O, Le Mee S, Pernelle C, Morfin R. 5Alpha-androstane-3beta, [71] Mathur C, Prasad VV, Raju VS, Welch M, Lieberman S. Steroids and their
7alpha, 17beta-triol and 5alpha-androstane-3beta, 7beta, 17beta-triol as conjugates in the mammalian brain. Proc Natl Acad Sci USA 1993;90:85–8.
substrates for the human 11beta-hydroxysteroid dehydrogenase type 1. [72] Liere P, Akwa Y, Weill Engerer S, Eychenne B, Pianos A, Robel P, et al.
Steroids 2007;72(13):855–64. Validation of an analytical procedure to measure trace amounts of
[44] Akwa Y, Morfin R, Robel P, Baulieu EE. Neurosteroids metabolism: 7a- neurosteroids in brain tissue by gas chromatography–mass spectrometry. J
hydroxylation of dehydroepiandrosterone and pregnenolone by rat brain Chromatogr B Biomed Sci Appl 2000;739:301–12.
microsomes. Biochem J 1992;288:959–64. [73] Li A, May MP, Bigelow JC. An LC/MS method for the quantitative
[45] Morfin R, Courchay G. Pregnenolone and dehydroepiandrosterone as determination of 7alpha-OH DHEA and 7beta-OH DHEA: an application for
precursors of active 7-hydroxylated metabolites which increase the the study of the metabolism of DHEA in rat brain. Biomed Chromatogr
immune response in mice. J Steroid Biochem Mol Biol 1994;50:91–100. 2010;24(8):833–7.
[46] Chalbot S, Trap C, Monin JP, Morfin R. Use of bioconversion for the [74] Tomlinson JW, Bujalska I, Stewart PM, Cooper MS. The role of 11 beta-
preparation of [4–14C]-labeled 7alpha- and 7beta-hydroxylated derivatives hydroxysteroid dehydrogenase in central obesity and osteoporosis. Endocr
of dehydroepiandrosterone and epiandrosterone. Steroids 2002;67:1121–7. Res 2000;26(4):711–22.
[47] Chalbot S, Morfin R. Neurosteroids: metabolism in human intestine [75] Muller C, Hennebert O, Morfin R. The native anti-glucocorticoid paradigm. J
microsomes. Steroids 2005;70:319–26. Steroid Biochem Mol Biol 2006;100:95–105.
[48] Inai Y, Nagai K, Ukena K, Oishi T, Tsutsui K. Seasonal changes in neurosteroid [76] Doostzadeh J, Urban P, Pompon D, Morfin R. Pregnenolone-7 beta-
concentrations in the amphibian brain and environmental factors regulating hydroxylating activities of yeast-expressed mouse cytochrome P450-1A1
their changes. Brain Res 2003;959:214–25. and mouse-tissue microsomes. Eur J Biochem 1996;242(3):641–7.
24 L.E. Kihel / Steroids 77 (2012) 10–26

[77] Kim SB, Hill M, Kwak YT, Hampl R, Jo DH, Morfin R. Neurosteroids: [101] Makrantonaki E, Schönknecht P, Hossini AM, Kaiser E, Katsouli MM, Adjaye J,
cerebrospinal fluid levels for Alzheimer’s disease and vascular dementia Schröder J, Zouboulis CC. Skin and brain age together: the role of hormones in
diagnostics. J Clin Endocrinol Metab 2003;88:5199–206. the ageing process. Exp Gerontol 2010;45(10):801–13.
[78] Naylor JC, Hulette CM, Steffens DC, Shampine LJ, Ervin JF, Payne VM, et al. [102] Qiu C, De Ronchi D, Fratiglioni L. The epidemiology of the dementias: an
Cerebrospinal fluid dehydroepiandrosterone levels are correlated with brain update. Curr Opin Psychiatry 2007;20:380–5.
dehydroepiandrosterone levels, elevated in Alzheimer’s disease, and related [103] Schonknecht P, Pantel J, Kruse A, Schroder J. Prevalence and natural course of
to neuropathological disease stage. J Clin Endocrinol Metab aging-associated cognitive decline in a population-based sample of young-
2008;93(8):3173–8. old subjects. Am J psychiatry 2005;162(11):2071–7.
[79] Starka L, Hill M, Kancheva R, Novak Z, Chrastina J, Pohanka M, et al. 7- [104] Schwartz AG, Pashko LL. Dehydroepiandrosterone, glucose-6-phosphate
Hydroxylated derivatives of dehydroepiandrosterone in the human dehydrogenase, and longevity. Ageing Res Rev 2004;3:171–87.
ventricular cerebrospinal fluid. Neuro Endocrinol Lett 2009;30(3):368–72. [105] Mamelak M. Alzheimer’s disease, oxidative stress and
[80] Morfin R. Involvement of steroids and cytochromes P450 species in the gammahydroxybutyrate. Neurobiol Aging 2007;28:1340–60.
triggering of immune defense. J Steroid Biochem Mol Biol 2002;80:273–90. [106] Chalbot S, Morfin R. Dehydroepiandrosterone metabolites and their
[81] Trincal M, Loeper D, Pompon D, Hauw JJ, Morfin R. DHEA metabolism in the interactions in humans. Drug Metabol Drug Interact. 2006;22:1–23.
brain production and effects of the 7a-hydroxylated derivative. In: Morfin R, [107] Yau JL, Rasmuson S, Andrew R, Graham M, Noble J, Olsson T, et al.
editor, DHEA and the brain. London: E-publishing Taylor and Francis; 2002. Dehydroepiandrosterone 7-hydroxylase CYP7B: predominant expression in
pp. 117–128. primate hippocampus and reduced expression in Alzheimer disease.
[82] Gordon G, Mackow MC, Levy HR. On the mechanism of interaction of steroids Neuroscience 2003;121:307–14.
with human glucose 6-phosphate dehydrogenase. Arch Biochem Biophys [108] Schumacher M, Weill-Engerer S, Liere P, Robert F, Franklin RJ, Garcia-Segura
1995;318:25–9. LM, et al. Steroid hormones and neurosteroids in normal and pathological
[83] Huynh PN, Loria RM. Contrasting effects of alpha- and beta-androstenediol on aging of the nervous system. Prog Neurobiol 2003;71(1):3–29.
oncogenic myeloid cell lines in vitro. J Leukoc Biol 1997;62:258–67. [109] Tagawa N, Sugimoto Y, Yamada J, Kobayashi Y. Strain differences of
[84] Graf MR, Jia W, Loria RM. The neuro-steroid, 3beta androstene 17alpha diol neurosteroid levels in mouse brain. Steroids 2006;71(9):776–84.
exhibits potent anti-tumour effects on human malignant glioma and [110] Pélissier MA, Trap C, Malewiak MI, Morfin R. Antioxidant effects of
lymphoma cells through different programmed cell death pathways. Br J dehydroepiandrosterone and 7a-hydroxy-dehydroepiandrosterone in the
Cancer 2007;97:619–27. rat colon, intestine and liver. Steroids 2004;69(2):137–44.
[85] Graf MR, Jia W, Johnson RS, Dent P, Mitchell C, Loria RM. Autophagy and the [111] Pélissier MA, Muller C, Hill M, Morfin R. Protection against dextran sodium
functional roles of Atg5 and beclin-1 in the anti-tumor effects of 3b- sulfate-induced colitis by dehydroepiandrosterone and 7alpha-hydroxy-
androstene 17a-diol neuro-steroid on malignant glioma cells. J Steroid dehydroepiandrosterone in the rat. Steroids 2006;71(3):240–8.
Biochem Mol Biol 2009;115(3–5):137–45. [112] Hennebert O, Pelissier MA, Le Mée S, Wülfert E, Morfin R. Anti-inflammatory
[86] Loria RM. Immune up-regulation and tumor apoptosis by androstene effects and changes in prostaglandin patterns induced by 7b-hydroxy-
steroids. Steroids 2002;67:953–66. epiandrosterone in rats with colitis. J Steroid Biochem Mol Biol
[87] Jia W, Loria RM, Park MA, Yacoub A, Dent P, Graf MR. The neuro-steroid, 5- 2008;110:255–62.
androstene 3b, 17a diol; induces endoplasmic reticulum stress and [113] Davidson J, Wulfert E, Rotondo D. 7beta-hydroxy-epiandrosterone
autophagy through PERK/eIF2a signaling in malignant glioma cells and modulation of 15-deoxy-delta12, 14-prostaglandin J2, prostaglandin D2
transformed fibroblasts. Int J Biochem Cell Biol 2010;42(12):2019–29. and prostaglandin E2 production from human mononuclear cells. J Steroid
[88] Graf MR, Jia W, Lewbart ML, Loria RM. The anti-tumor effects of androstene Biochem Mol Biol 2008;112(4–5):220–7.
steroids exhibit a strict structure-activity relationship dependent upon the [114] Le Mée S, Hennebert O, Ferrec C, Wülfert E, Morfin R. 7b-Hydroxy-
orientation of the hydroxyl group on carbon-17. Chem Biol Drug Des epiandrosterone-mediated regulation of the prostaglandin synthesis
2009;74(6):625–9. pathway in human peripheral blood monocytes. Steroids 2008;73:1148–59.
[89] Jellinck PH, Lee SJ, McEwen BS. Metabolism of dehydroepiandrosterone by rat [115] Schmidt M, Kreutz M, Loffler G, Scholmerich J, Straub RH. Conversion of
hippocampal cells in culture: possible role of aromatization and 7- dehydroepiandrosterone to downstream steroid hormones in macrophages. J
hydroxylation in neuroprotection. J Steroid Biochem Mol Biol Endocrinol 2000;164:161–9.
2001;78(4):313–7. [116] Dulos J, Verbraak E, Bagchus WM, Boots AM, Kaptein A. Severity of murine
[90] Jellinck PH, Croft G, McEwen BS, Gottfried-Blackmore A, Jones G, Byford V, collagen-induced arthritis correlates with increased CYP7B activity:
Bulloch K. Metabolism of dehydroepiandrosterone by rodent brain cell lines: enhancement of dehydroepiandrosterone metabolism by interleukin-1beta.
relationship between 7-hydroxylation and aromatization. J Steroid Biochem Arthritis Rheum 2004;50(10):3346–53.
Mol Biol 2005;93(1):81–6. [117] Dulos J, van der Vleuten MA, Kavelaars A, Heijnen CJ, Boots AM. CYP7B
[91] Jellinck PH, Kaufmann M, Gottfried-Blackmore A, Croft G, Byford V, McEwen expression and activity in fibroblast-like synoviocytes from patients with
BS, Jones G, Bulloch K. Dehydroepiandrosterone (DHEA) metabolism in the rheumatoid arthritis: regulation by proinflammatory cytokines. Arthritis
brain: identification by liquid chromatography/mass spectrometry of the Rheum 2005;52(3):770–8.
delta-4-isomer of DHEA and related steroids formed from androstenedione [118] Dulos J, Boots AH. DHEA metabolism in arthritis: a role for the p450 enzyme
by mouse BV2 microglia. J Steroid Biochem Mol Biol 2006;98(1):41–7. Cyp7b at the immune-endocrine crossroad. Ann NY Acad Sci
[92] Migues PV, Johnston AN, Rose SP. Dehydroepiandosterone and its sulphate 2006;1069:401–13.
enhance memory retention in day-old chicks. Neuroscience [119] Hill M, Havlíková H, Vrbíková J, Kancheva R, Kancheva L, Pouzar V, Cerný I,
2002;109:243–51. Stárka L. The identification and simultaneous quantification of 7-
[93] Jellinck PH, Kaufmann M, Gottfried-Blackmore A, McEwen BS, Jones G, hydroxylated metabolites of pregnenolone, dehydroepiandrosterone, 3beta,
Bulloch K. Selective conversion by microglia of dehydroepiandrosterone to 5- 17betaandrostenediol, and testosterone in human serum using gas
androstenediol-A steroid with inherent estrogenic properties. J Steroid chromatography-mass spectrometry. J Steroid Biochem Mol Biol
Biochem Mol Biol 2007;107(3–5):156–62. 2005;96:187–200.
[94] Sujkovic E, Mileusnic R, Fry JP, Rose SP. Temporal effects of [120] Offner H, Zamora A, Drought H, Matejuk A, Auci DL, Morgan EE, Vandenbark
dehydroepiandrosterone sulfate on memory formation in day-old chicks. AA, Reading CL. A synthetic androstene derivative and a natural androstene
Neuroscience 2007;148(2):375–84. metabolite inhibit relapsing-remitting EAE. J Neuroimmunol
[95] Aldred S, Mecocci P. Decreased dehydroepiandrosterone (DHEA) and 2002;130:128–39.
dehydroepiandrosterone sulfate (DHEAS) concentrations in plasma of [121] Auci DL, Ahlem C, Li M, Trauger R, Dowding C, Paillard F, Frincke J, Reading CL.
Alzheimer’s disease (AD) patients. Arch Gerontol Geriatr 2010;51(1):e16–8. The immunobiology and therapeutic potential of androstene hormones and
[96] Shi J, Schulze S, Lardy HA. The effect of 7-oxo-DHEA acetate on memory in their synthetic derivatives: novel anti-inflammatory and immune regulating
young and old C57BL/6 mice. Steroids 2000;65(3):124–9. steroid hormones. Mod Asp Immunobiol 2003;3:64–70.
[97] Yau JL, Noble J, Graham M, Seckl JR. Central administration of a cytochrome [122] Marcu AC, Kielar ND, Paccione KE, Barbee RW, Carter H, Ivatury RR,
P450–7B product 7 alpha-hydroxypregnenolone improves spatial memory Diegelmann RF, Ward KR, Loria RM. Androstenetriol improves survival in a
retention in cognitively impaired aged rats. J Neurosci 2006;26(43): rodent model of traumatic shock. Resuscitation 2006;71:379–86.
11034–40. [123] Offner H, Firestein GS, Boyle DL, Pieters R, Frincke JM, Garsd A, White SK,
[98] Wolf OT, Neumann O, Hellhammer DH, Geiben AC, Strasburger CJ, Reading CL, Auci DL. An orally bioavailable synthetic analog of an active
Dressendorfer RA, et al. Effects of a two-week physiological dehydroepiandrosterone metabolite reduces established disease in rodent
dehydroepiandrosterone substitution on cognitive performance and well- models of rheumatoid arthritis. J Pharmacol Exp Ther 2009;329(3):1100–9.
being in healthy elderly women and men. J Clin Endocrinol Metab [124] Schmidt M, Naumann H, Weidler C, Schellenberg M, Anders S, Straub RH.
1997;82:2363–7. Inflammation and sex hormone metabolism. Ann NY Acad Sci
[99] Yamada S, Akishita M, Fukai S, Ogawa S, Yamaguchi K, Matsuyama J, Kozaki K, 2006;1069:236–46.
Toba K, Ouchi Y. Effects of dehydroepiandrosterone supplementation on [125] Auci DL, Reading CL, Frincke JM. 7-Hydroxy androstene steroids and a novel
cognitive function and activities of daily living in older women with mild to synthetic analogue with reduced side effects as a potential agent to treat
moderate cognitive impairment. Geriatr Gerontol Int 2010;10(4):280–7. autoimmune diseases. Autoimmun Rev 2009;8:369–72.
[100] Alhaj HA, Massey AE, McAllister-Williams RH. Effects of DHEA administration [126] Canning MO, Grotenhuis K, de Wit HJ, Drexhage HA. Opposing effects of
on episodic memory, cortisol and mood in healthy young men: a double-blind, dehydroepiandrosterone and dexamethasone on the generation of
placebo-controlled study. Psychopharmacology (Berl) 2006;188(4):541–51. monocyte-derived dendritic cells. Eur J Endocrinol 2000;143:687–95.
L.E. Kihel / Steroids 77 (2012) 10–26 25

[127] Harding G, Mak YT, Evans B, Cheung J, Macdonald D, et al. The effects of [153] Shin MH, Rhie GE, Park CH, Kim KH, Cho KH, Eun HC, et al. Modulation of
dexamethasone and dehydroepiandrosterone (DHEA) on cytokines and collagen metabolism by the topical application of dehydroepiandrosterone to
receptor expression in a human osteoblastic cell line: Potential steroid- human skin. J Invest Dermatol 2005;124:315–23.
sparing role for DHEA. Cytokine 2006;36:57–68. [154] Calvo E, Luu-The V, Morissette J, Martel C, Labrie C, Bernard B, Bernerd F,
[128] Wang L, Wang YD, Wang WJ, Zhu Y, Li DJ. Dehydroepiandrosterone improves Deloche C, Chaussade V, Leclaire J, Labrie F. Pangenomic changes induced by
murine osteoblast growth and bone tissue morphometry via mitogen- DHEA in the skin of postmenopausal women. J Steroid Biochem Mol Biol
activated protein kinase signaling pathway independent of either androgen 2008;112(4–5):186–93.
receptor or estrogen receptor. J Mol Endocrinol 2007;38:467–79. [155] Luu-the V, Ferraris C, Duche D, Bélanger P, Leclaire J, Labrie F. Steroid
[129] Hartkamp A, Geenen R, Godaert GL, Bijl M, Bijlsma JW, Derksen RH. The effect metabolism and profile of steroidogenic gene expression in Episkin: high
of dehydroepiandrosterone on lumbar spine bone mineral density in patients similarity with human epidermis. J Steroid Biochem Mol Biol 2007;107(1–
with quiescent systemic lupus erythematosus. Arthritis Rheum 2):30–6.
2004;50(11):3591–5. [156] Mizushima Y, Wang P, Jarrar D, Cioffi WG, Bland KI, Chaudry IH. Estradiol
[130] Sánchez-Guerrero J, Fragoso-Loyo HE, Neuwelt CM, Wallace DJ, Ginzler EM, administration after trauma-hemorrhage improves cardiovascular and
Sherrer YR, et al. Effects of prasterone on bone mineral density in women hepatocellular functions in male animals. Ann Surg 2000;232:673–9.
with active systemic lupus erythematosus receiving chronic glucocorticoid [157] Ba ZF, Wang P, Koo DJ, Cioffi WG, Bland KI, Chaudry IH. Alterations in tissue
therapy. J Rheumatol 2008;35(8):1567–75. oxygen consumption and extraction after trauma and hemorrhagic shock.
[131] Von Mühlen D, Laughlin GA, Kritz-Silverstein D, Bergstrom J, Bettencourt R. Crit Care Med 2000;28:2837–42.
Effect of dehydroepiandrosterone supplementation on bone mineral density, [158] Ba ZF, Kuebler JF, Rue III LW, Bland KI, Wang P, Chaudry IH. Gender dimorphic
bone markers, and body composition in older adults: the DAWN trial. tissue perfusion response after acute hemorrhage and resuscitation: role of
Osteoporos Int 2008;19(5):699–707. vascular endothelial cell function. Am J Physiol Heart Circ Physiol
[132] Urban NH, Chamberlin B, Ramage S, Roberts Z, Loria RM, Beckman MJ. Effects 2003;284:H2162–9.
of a/b-androstenediol immune regulating hormones on bone remodeling and [159] Shimizu T, Choudhry MA, Szalay L, Rue III LW, Bland KI, Chaudry IH. Salutary
apoptosis in osteoblasts. J Steroid Biochem Mol Biol 2008;110(3–5):223–9. effects of androstenediol on cardiac function and splanchnic perfusion after
[133] Boyce BF, Xing L. Biology of RANK, RANKL, and osteoprotegerin. Arthritis Res trauma-hemorrhage. Am J Physiol Regul Integr Comp Physiol
Ther 2007;9(Suppl 1):S1. 2004;287(2):R386–90.
[134] Malik AK, Khaldoyanidi S, Auci DL, Miller SC, Ahlem CN, Reading CL, Page T, [160] Shimizu T, Szalay L, Choudhry MA, Schwacha MG, Rue III LW, Bland KI,
Frincke JM. 5-Androstene-3b, 7b, 17b-triol (b-AET) slows thermal injury Chaudry IH. Mechanism of salutary effects of androstenediol on hepatic
induced osteopenia in mice. relation to aging and osteoporosis. PLoS One function after trauma-hemorrhage: role of endothelial and inducible nitric
2010;5(10):e13566. oxide synthase. Am J Physiol Gastrointest Liver Physiol
[135] Su CY, Lardy H. Induction of hepatic mitochondrial glycerophosphate 2005;288(2):G244–50.
dehydrogenase in rats by dehydroepiandrosterone. J Biochem [161] Nilsson S, Mäkelä S, Treuter E, Tujague M, Thomsen J, Andersson G, et al.
1991;110(2):207–13. Mechanisms of estrogen action. Physiol Rev 2001;81:1535–65.
[136] Bobyleva V, Kneer N, Bellei M, Batelli D, Lardy H. Concerning the mechanism [162] Matthews J, Gustafsson JÅ. Estrogen signaling: a subtle balance between ERa
of increased thermogenesis in rats treated with dehydroepiandrosterone. J and ERb. Mol Interv 2003;3:281–92.
Bionerg Biomembr 1993;25(3):313–21. [163] Maggiolini M, Donze O, Jeannin E, Ando S, Picard D. Adrenal androgens
[137] Marwah P, Marwah A, Kneer N, Lardy H. Ergosteroids IV: synthesis and stimulate the proliferation of breast cancer cells as direct activators of
biological activity of steroid glucuronosides, ethers, and alkylcarbonates. estrogen receptor alpha. Cancer Res 1999;59:4864–9.
Steroids 2001;66(7):581–95. [164] Chen F, Knecht K, Birzin E, Fisher J, Wilkinson H, Mojena M, et al. Direct
[138] Ihler G, Chami-Stemman H. 7-Oxo-DHEA and Raynaud’s phenomenon. Med agonist/antagonist functions of dehydroepiandrosterone (DHEA).
Hypotheses 2003;60:391–7. Endocrinology 2005;146:4568–96.
[139] Lardy H, Partridge B, Kneer N, Wei Y. Ergosteroids: induction of thermogenic [165] Weihua Z, Lathe R, Warner M, Gustafsson JÅ. An endocrine pathway in the
enzymes in liver of rats treated with steroids derived from prostate, ERb, AR, 5a-androstane-3b, 17b-diol, and CYP7B1, regulates
dehydroepiandrosterone. Proc Natl Acad Sci USA 1995;92(14):6617–9. prostate growth. Proc Natl Acad Sci USA 2002;99:13589–94.
[140] Bobyleva V, Bellei M, Kneer N, Lardy H. The effects of the ergosteroid 7-oxo- [166] Martin C, Ross M, Chapman KE, Andrew R, Bollina P, Seckl JR, Habib FK. CYP7B
dehydroepiandrosterone on mitochondrial membrane potential: possible generates a selective estrogen receptor b agonist in human prostate. J Clin
relationship to thermogenesis. Arch Biochem Biophys 1997;341(1):122–8. Endocrinol Metab 2004;89:2928–35.
[141] Tagawa N, Takano T, Fukata S, Kuma K, Tada H, Izumi Y, et al. Serum [167] Umetani M, Domoto H, Gormley AK, Yuhanna IS, Cummins CL, Javitt NB, et al.
concentrations of androstenediol and androstenediol sulfate in patients with 27-Hydroxycholesterol is an endogenous SERM that inhibits the
hyperthyroidism and hypothyroidism. Endocr J 2001;48(3):345–54. cardiovascular effects of estrogen. Nat Med 2007;13:1185–92.
[142] Sulcová J, Hampl R, Hill M, Stárka L, Novácek A. Delayed effects of short-term [168] DuSell CD, Umetani M, Shaul PW, Mangelsdorf DJ, McDonnell DP. 27-
transdermal application of 7-oxo-dehydroepiandrosterone on its Hydroxycholesterol is an endogenous selective estrogen receptor modulator.
metabolites, some hormonal steroids and relevant proteohormones in Mol Endocrinol 2008;22:65–77.
healthy male volunteers. Clin Chem Lab Med 2005;43(2):221–7. [169] Pettersson H, Lundqvist J, Norlin M. Effects of CYP7B1-mediated catalysis on
[143] Hampl R, Sulcová J, Bílek R, Hill M. How short-term transdermal treatment of estrogen receptor activation. Biochim Biophys Acta 2010;1801(9):1090–7.
men with 7-oxo-dehydroepiandrosterone influence thyroid function. Physiol [170] Negro-Vilar A. Selective androgen receptor modulators (SARMs): a novel
Res 2006;55(1):49–54. approach to androgen therapy for the new millennium. J Clin Endocrinol
[144] Drbalová K, Matucha P, Matejková-Behanová M, Bílek R, Kríz L, Kazihnitková Metab 1999;84:3459–62.
H, Hampl R. Immunoprotective steroids and SHBG in non-treated [171] Miyamoto H, Yeh S, Lardy H, Messing E, Chang C. D5-Androstenediol is a
hypothyroidism and their relationship to autoimmune thyroid disorders. natural hormone with androgenic activity in human prostate cancer cells.
Physiol Res 2008;57:S119–25. Proc Natl Acad Sci USA 1998;95(19):11083–8.
[145] Ntambi JM, Kim YC. Adipocyte differentiation and gene expression. J Nutr [172] Chang HC, Miyamoto H, Marwah P, Lardy H, Yeh S, Huang KE, Chang C.
2000;130(12):3122S–6S. Suppression of Delta(5)-androstenediol-induced androgen receptor
[146] Rice SP, Zhang L, Grennan-Jones F, Agarwal N, Lewis MD, Rees DA, Ludgate M. transactivation by selective steroids in human prostate cancer cells. Proc
Dehydroepiandrosterone (DHEA) treatment in vitro inhibits adipogenesis in Natl Acad Sci USA 1999;96(20):11173–7.
human omental but not subcutaneous adipose tissue. Mol Cell Endocrinol [173] Marwah P, Marwah A, Lardy HA, Miyamoto H, Chang C. C19-Steroids as
2010;320(1–2):51–7. androgen receptor modulators: Design, discovery, and structure-activity
[147] Gomez FE, Miyazaki M, Kim YC, Marwah P, Lardy HA, Ntambi JM, Fox BG. relationship of new steroidal androgen receptor antagonists. Bioorg Med
Molecular differences caused by differentiation of 3T3–L1 preadipocytes in Chem 2006;14(17):5933–47.
the presence of either dehydroepiandrosterone (DHEA) or 7-oxo-DHEA. [174] Mo Q, Lu SF, Simon NG. Dehydroepiandrosterone and its metabolites:
Biochemistry 2002;41(17):5473–82. differential effects on androgen receptor trafficking and transcriptional
[148] Marwah A, Gomez FE, Marwah P, Ntambi JM, Fox BG, Lardy H. Redox activity. J Steroid Biochem Mol Biol 2006;99(1):50–8.
reactions of dehydroepiandrosterone and its metabolites in differentiating [175] Pelletier G. Expression of steroidogenic enzymes and sex-steroid receptors in
3T3–L1 adipocytes: A liquid chromatographic–mass spectrometric study. human prostate. Best Pract Res Clin Endocrinol Metab 2008;22:223–8.
Arch Biochem Biophys 2006;456(1):1–7. Erratum in: Arch Biochem Biophys [176] Luu-the V, Bélanger A, Labrie F. Androgen biosynthetic pathways in the
2007; 465(1):302. human prostate. Best Pract Res Clin Endocrinol Metab 2008;22(2):207–21.
[149] Chen W, Thiboutot D, Zouboulis CC. Cutaneous androgen metabolism: basic [177] Leskelä S, Honrado E, Montero-Conde C, Landa I, Cascón A, Letón R, et al.
research and clinical perspectives. J Invest Dermatol 2002;119:992–1007. Cytochrome P450 3A5 is highly expressed in normal prostate cells but absent
[150] Labrie F, Cusan L, Gomez JL, Martel C, Bérubé R, Bélanger P, et al. Changes in in prostate cancer. Endocr Relat Cancer 2007;14(3):645–54.
serum DHEA and eleven of its metabolites during 12-month percutaneous [178] Mitamura K, Nakagawa T, Shimada K, Namiki M, Koh E, Mizokami A, et al.
administration of DHEA. J Steroid Biochem Mol Biol 2008;110(1–2):1–9. Identification of dehydroepiandrosterone metabolites formed from human
[151] Labrie, Luu-the V, Labrie C, Pelletier G, El-Alfy M. Intracrinology and the skin. prostate homogenate using liquid chromatography–mass spectrometry and
Horm Res 2000;54:218–29. gas chromatography–mass spectrometry. J Chromatogr A 2002; 961:97–105.
[152] Lee KS, Oh KY, Kim BC. Effects of dehydroepiandrosterone on collagen and [179] Mohler JL, Gregory CW, Ford OH, Kim D, Weaver CM, Petrusz P, et al. The
collagenase gene expression by skin fibroblasts in culture. J Dermatol Sci androgen axis in recurrent prostate cancer. Clin Cancer Res
2000;23:103–10. 2004;10(1):440–8.
26 L.E. Kihel / Steroids 77 (2012) 10–26

[180] Arai S, Miyashiro Y, Shibata Y, Tomaru Y, Kobayashi M, Honma S, Suzuki K. [185] Risbridger GP, Ellem SJ, McPherson SJ. Estrogen action on the prostate gland:
Effect of castration monotherapy on the levels of adrenal androgens in a critical mix of endocrine and paracrine signaling. J Mol Endocrinol
cancerous prostatic tissues. Steroids 2011;76(3):301–8. 2007;39:183–8.
[181] Widstrom RL, Dillon JS. Is there a receptor for dehydroepiandrosterone or [186] Carruba G. Estrogen and prostate cancer: an eclipsed truth in an androgen-
dehydroepiandrosterone sulfate? Semin Reprod Med 2004;22:289–98. dominated scenario. J Cell Biochem 2007;102:899–911.
[182] Martin C, Ross M, Chapman KE, Andrew R, Bollina P, Seckl JR, Habib FK. CYP7B [187] Hsing AW, Chu LW, Stanczyk FZ. Androgen and prostate cancer: is the
generates a selective estrogen receptor beta agonist in human prostate. J Clin hypothesis dead? Cancer Epidemiol Biomarkers Prev 2008;17:2525–30.
Endocrinol Metab 2004;89:2928–35. [188] Arnold JT, Blackman MR. Does DHEA exert direct effects on androgen and
[183] McPherson SJ, Ellem SJ, Simpson ER, Patchev V, Fritzemeier KH, Risbridger GP. estrogen receptors, and does it promote or prevent prostate cancer?
Essential role for estrogen receptor beta in stromal-epithelial regulation of Endocrinology 2005;146:4565–7.
prostatic hyperplasia. Endocrinology 2007;148:566–74. [189] Arnold JT. DHEA metabolism in prostate: For better or worse? Mol Cell
[184] McPherson SJ, Ellem SJ, Risbridger GP. Estrogen-regulated development and Endocrinol 2009;301(1–2):83–8.
differentiation of the prostate. Differentiation 2008;76:660–70.

You might also like