Sie sind auf Seite 1von 11

Molecular and Cellular Endocrinology 215 (2004) 149–159

Review
Modulation of aldosterone and cortisol synthesis on the molecular level
Michael Lisurek, Rita Bernhardt∗
Universität des Saarlandes, FR 8.8 Biochemie, Postfach 151150, 66041 Saarbrücken, Germany

Abstract

CYP11B1 and the closely related CYP11B2 are involved in the production of adrenal steroid hormones. Although in human their primary
structure is 93% identical they are involved in the biosynthesis of functionally diverse products, such as glucocorticoids and mineralocorticoids,
respectively. In contrast, bovine CYP11B1 combines both activities in one single enzyme. The CYP11B family belongs to class I cytochromes
P450 that have been described in bacteria and mitochondria and receive their electrons from a low molecular weight iron sulphur protein
which is reduced by a NADPH-dependent FAD-containing reductase.
In this review, we summarise the current knowledge on the modulation of aldosterone and cortisol synthesis by transcriptional regulation,
on the molecular level as consequence of mutations found in patients suffering from steroid hormone-related diseases as well as introduced by
site-directed mutagenesis and as consequence of protein–protein interaction with both CYP11A1 and the natural redox partner adrenodoxin.
© 2003 Elsevier Ireland Ltd. All rights reserved.

Keywords: CYP11B1; CYP11B2; Cytochrome P450; 11␤-Hydroxylase; Aldosterone synthase; Glucocorticoids; Mineralocorticoids

1. Introduction strated that hyperaldosteronism may be accompanied by


hypertension and heart disease (see below).
The most common reaction catalysed by cytochromes CYP11B enzymes of other species have also been stud-
P450 is hydroxylation of organic compounds, but they are ied, and it could be shown that also in the adrenal cor-
also capable of performing a wide spectrum of different re- tex of rat (Matsukawa et al., 1990), mouse (Domalik et al.,
actions including N-oxidation, N-, S-, and O-dealkylation, 1991), guinea pig (Bülow et al., 1996; Bülow and Bernhardt,
sulfoxidation, epoxidation, peroxidation, deamination, 2002), baboon (Hampf et al., 1996; Swart et al., 2000)
desulfuration, dehalogenation and N-oxide reduction (Ortiz and hamster (Veronneau et al., 1996), two distinct isoforms
De Montellano, 1995; Bernhardt, 1996). Cytochromes P450 are involved in the formation of either mineralo- or glu-
are key enzymes responsible for the metabolism of drugs cocorticoids. In contrast, in cows (Wada et al., 1985), pigs
and other xenobiotics and the anabolism of important en- (Yanagibashi et al., 1986), sheep (Boon et al., 1995) and
dogenous compounds like steroid hormones, bile acids or frogs (Nonaka et al., 1995) the synthesis of gluco- and
prostaglandins. mineralocorticoids is catalysed by a single enzyme, termed
In humans the final steps of cortisol and aldosterone CYP11B1. In rat a third CYP11B isoform is expressed
synthesis are performed by 11␤-hydroxylase (CYP11B1) with 11␤- and 18-hydroxylase activities, but no detectable
and aldosterone synthase (CYP11B2), respectively (Mornet 18-oxidase activity (Mellon et al., 1995).
et al., 1989; Kawamoto et al., 1990a,b). Cortisol is the main
glucocorticoid in humans. It regulates energy mobilisation,
stress response and is additionally involved in the immune 2. Aldosterone biosynthesis
response of the human body. Aldosterone is the most po-
tent natural human mineralocorticoid. It participates in the Human CYP11B1 and CYP11B2 catalyse the final steps
regulation of the salt and water household of the body and in gluco- and mineralocorticoid synthesis, respectively
thus in the regulation of blood pressure. It has been demon- (Fig. 1). CYP11B1 is expressed at high levels in the zona
fasciculata/reticularis of the adrenal cortex (Erdmann et al.,
∗ Corresponding author. Tel.: +49-681-302-4241; 1995a,b) and produces cortisol, the principal human glu-
fax: +49-681-302-4739. cocorticoid. In contrast, aldosterone secretion (Miller and
E-mail address: ritabern@rz.uni-sb.de (R. Bernhardt). Tyrell, 1995) and CYP11B2 expression (Pascoe et al., 1995)

0303-7207/$ – see front matter © 2003 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2003.11.008
150 M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159

Fig. 1. Reactions catalysed by human CYP11B1 and CYP11B2: (A) CYP11B1 catalyses the 11␤-hydroxylation of 11-deoxycortisol to cortisol; (B)
CYP11B2 converts 11-deoxycorticosterone via corticosterone and 18-hydroxycorticosterone to aldosterone.

take place at low levels in the zona glomerulosa of the have shown that the two intermediates leading to aldos-
adrenal cortex. This zonal distribution of expression has terone from 11-deoxycorticosterone are not released from
recently been further investigated with surgically removed the active site of the enzyme (Imai et al., 1998). This can
human adrenal gland cells, showing a higher concentration also be expected to be the case for human CYP11B2, since
of CYP11B1 in the zona fasciculata than in the zona reticu- it is known that the intermediates are poor substrates for
laris (Young et al., 2003). Cortisol is usually secreted 100- to human CYP11B2 (Denner et al., 1995).
1000-fold in excess over aldosterone, but this is not only due Both CYP11B1 and CYP11B2 expression have also been
to transcriptional regulation of these enzymes, but also to dif- detected in extra adrenal tissue such as rat brain (Ozaki et al.,
ferences in catalytic activities. The adrenal steroid hormones 1991; Erdmann et al., 1996; MacKenzie et al., 2000) and
are produced in multi-step pathways involving five differ- heart (Rudolph et al., 2000; Yoshimura et al., 2002). Even
ent cytochromes P450 and 3␤-HSD, a member of the short if their function in most of these tissue is not fully under-
chain dehydrogenase family (Fig. 2): CYP11A1 (side chain stood, it is known that chronic overproduction of aldosterone
cleavage enzyme), CYP17 (steroid 17␣-hydroxylase and leads to an increased blood volume and stimulates cardiac
17,20-lyase), CYP21 (steroid 21-hydroxylase), CYP11B1 hypertrophy, myocardial fibrosis and ventricular arrhythmia
(steroid 11␤-hydroxylase) and CYP11B2 (aldosterone syn- (Brilla, 2000; Lijnen and Petrov, 2000). In the heart, further
thase) (Bernhardt, 1996; Bureik et al., 2002a). In vitro pathological consequences such as endothelial dysfunction
reconstitution of the CYP11B1 electron transfer chain and tissue damage are observed by aldosterone dysregula-
showed that 11-deoxycorticosterone and, to a minor extent, tion (Takeda et al., 2000).
11-deoxycortisol are 11␤-hydroxylated (Kawamoto et al.,
1990a; Curnow et al., 1991; Denner et al., 1995; Fisher
et al., 2001). In contrast to this, human CYP11B2 exhibits 3. Differences and similarities between CYP11B1 and
a weak 11␤-hydroxylase activity towards 11-deoxycortisol, CYP11B2
but 11␤-hydroxylates 11-deoxycorticosterone to corticos-
terone and in addition, 18-hydroxylates and 18-oxidises Human CYP11B1 and CYP11B2 are both synthesised
it further to 18-hydroxycorticosterone and aldosterone, as 503 amino acids containing precursor proteins (Mornet
respectively (Fig. 1). CYP11B2 can also 18-hydroxylate et al., 1989; Kawamoto et al., 1990a), both containing a
and 18-oxidise cortisol. Bovine CYP11B1, producing both 24-residue N-terminal mitochondrial targeting sequence
gluco- and mineralocorticoids, can be purified from adreno- which is cleaved off after translocation into the mitochon-
cortical mitochondria (Akhrem et al., 1979). Kinetic studies drial matrix. In the mature enzymes, only 29 out of 479
M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159 151

Fig. 2. Steroid biosynthesis in the adrenal cortex. The diagram shows the reactions leading from cholesterol to cortisol, aldosterone and androstenedione
including the names of the corresponding enzymes and the organelles in which the reactions are carried out.

residues are not identical (Belkina et al., 2001). All of and CYP11B2 have apparent molecular masses of 50 and
these residues seem to be located outside of the generally 48.5 kDa, respectively, and are bound to the inner mito-
accepted substrate recognition sites (Gotoh, 1992) and are chondrial membrane by as yet undefined protein segments
spread throughout the protein (Fig. 3). Human CYP11B1 (Ogishima et al., 1991).
Understanding the structure–function relationships
of CYP11B enzymes requires information about their
three-dimensional structure. Protein structure determina-
tion by X-ray diffraction is often problematic in case of
membrane-bound proteins such as CYP11B1 and CYP11B2,
and NMR structure determination is restricted to smaller
proteins. Due to these reasons, no structure of a mitochon-
drial cytochrome P450 has been experimentally resolved so
far. Until recently only the structures of several bacterial
cytochromes P450 and that of a single microsomal P450
(CYP2C5), solubilised by truncation and site-directed mu-
tagenesis (Cosme and Johnson, 2000) has been determined
experimentally (Williams et al., 2000). While this paper was
in preparation, one more structure of a membrane bound
cytochromes P450 (CYP2C9) became available (Williams
et al., 2003). Analysis of the structures revealed a conserved
structural fold. Therefore, homology modelling studies of
human CYP11B1 and CYP11B2 have been performed. The
Fig. 3. Distribution of the amino acids differing between human CYP11B1 models have been evaluated and used to explain the signif-
and CYP11B2. The C␣-positions of the exchanged amino acid are labelled
and shown as black balls on the ribbon structure of the homology model
icance of a number of residues that were identified either
of CYP11B2 (Belkina et al., 2001). The heme is represented as grey stick by mutagenesis studies or found in patients (Belkina et al.,
model. Amino acids 301, 302 and 320 can be found in the I-helix. 2001).
152 M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159

the adrenocorticotropic hormone (ACTH) (Waterman and


Simson, 1989), and the expression of the CYP11B2 gene
is mainly controled by the renin–angiotensin system (Quinn
and Williams, 1988). Angiotensin II and potassium increase
the production levels of CYP11B2, but aldosterone synthe-
sis is also influenced by other factors such as sodium status
or the atrial natriuretic peptide (ANP).
A -344 C/T polymorphism in the promoter region of hu-
man CYP11B2 has been associated with transcriptional reg-
ulation of this gene (Kupari et al., 1998; Lim et al., 2002),
but so far experimental evidence have not underlined this as-
sumption leading to a controversial discussion of the matter
(Schunkert et al., 1999; Hautanen et al., 1999; Delles et al.,
2001; Connell et al., 2003).
The CYP11B1 and CYP11B2 genes are arranged
tandemly on chromosome 8, approximately 40 kb apart
from each other (Chua et al., 1987; Wagner et al., 1991).
Recent studies have mapped human CYP11B2 to the cy-
Fig. 4. Superposition of the ribbon structures of the homology models togenic band 8q24.3 (Taymans et al., 1998). Chimeric
of human CYP11B1 (black) and CYP11B2 (grey) (Belkina et al., 2001). enzymes of CYP11B1 and CYP11B2, derived by unequal
Helices are labelled in the letter code. While the overall structure is similar, crossing over, lead to severe consequences as displayed in
the position of the hemes in both proteins is different. The orientation of
the model is essentially as shown in Fig. 3.
patients suffering from familial hyperaldosteronism type I
(also called glucocorticoid remediable hyperaldosteronism)
(Lifton et al., 1992; MacConnachie et al., 1998; Seeman
These models suggest that the main difference between
et al., 1999). The chimeric genes are composed of the
the two proteins is the position of the heme. An angle of
5 -coding sequence and regulatory elements of CYP11B1
approximately 20◦ between the hemes of the two models
fused to the 3 -coding sequence of CYP11B2 and exhibit
has been observed, apparently dependent on the interaction
aldosterone synthesis under the regulatory control of the
of side-chains forming the heme environment and the ori-
ACTH-sensitive promoter, thus leading to hypersecretion of
entation of its binding loop (Fig. 4). In case of CYP11B1,
aldosterone. Administration of dexamethasone (or any other
one heme propionate group forms a hydrogen bond with
glucocorticoid) suppresses ACTH production and results in
Arg448 while the second interacts with Arg384, whereas
corrections of the biochemical anomaly. Patients suffering
in CYP11B2 both heme propionate groups are involved
from familial hyperaldosteronism type I carrying genes
in hydrogen bond interactions with Arg448. Both, Arg384
consisting of the first four exons of CYP11B1 and the last
and Arg448, have been found to be mutated in CYP11B1
five exons of CYP11B2 have been found. Transfection ex-
of patients suffering from congenital adrenal hyperplasia
periments with chimeras, where only the last four exons of
(CAH) (Curnow et al., 1993; Nakagawa et al., 1995; Geley
CYP11B1 were replaced by the corresponding ones found
et al., 1996; White et al., 1991) (see below). All known
in CYP11B2, showed that these constructs had activities
mutations in positions 384 and 448 led to a complete loss
similar to cells transfected with CYP11B1 and no aldos-
of enzyme activity, most probably due to destabilisation of
terone formation could be detected (Pascoe et al., 1992b).
the holoprotein. As a consequence of the different hydrogen
These findings suggest that exon 5 of CYP11B2 is essential
bonding network around Arg384, Arg448 and the heme
in conferring aldosterone synthase activity to CYP11B1. A
propionates, the active site of CYP11B2 is predicted to be
chimera consisting of the promotor and exon 1 to exon 4
smaller than that of CYP11B1. The larger active site found
of CYP11B2 and exon 5 to exon 9 of CYP11B1 was found
in the CYP11B1 model correlates with the fact that the
in a patient suffering from congenital adrenal hyperplasia,
natural substrate of CYP11B1 (11-deoxycortisol) is larger
due to insufficient expression of this gene resulting from
than that of CYP11B2 (11-deoxycorticosterone) due to the
the replacement of the strong ACTH-inducible promoter by
presence of an additional 17␣-hydroxy group (Fig. 1).
the regulatory elements of CYP11B2 (Hampf et al., 2001).

4. Regulation on the level of transcription


5. Regulation on the molecular level
The coding sequences of the CYP11B1 and CYP11B2
genes are highly homologous, whereas their promoter re- 5.1. Activity modulation by the primary structure
gions are strikingly different, emphasising the fact that they
are regulated differently (Lifton et al., 2001; White, 2001; Valuable clues for the identification of important residues
Ferrari, 2002). Usually CYP11B1 activity is regulated by in human CYP11B enzymes came from the analysis of
M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159 153

Table 1
List of published missense mutations in the human CYP11B1 gene
Mutation Patient Exon Putative 3D location Effect in vitro Reference

Pro42Ser Female girl 1 A-helix 15% reduced 11␤-hydroxylase activity Joehrer et al. (1997)
Val129Met Caucasian, male 2 C-helix Complete loss of 11␤-hydroxylase activity Geley et al. (1996)
Asn133His Female girl 3 C-helix 17% reduced 11␤-hydroxylase activity Joehrer et al. (1997)
Thr318Met Yemenite, female 5 I-helix, active site Complete loss of 11␤-hydroxylase activity Curnow et al. (1993)
Thr319Met Female girl 6 I-helix, active site 37% reduced 11␤-hydroxylase activity Joehrer et al. (1997)
Ala331Val Caucasian, female 6 I-helix Complete loss of 11␤-hydroxylase activity Geley et al. (1996)
Glu371Gly Caucasian, female 6 K-helix Complete loss of 11␤-hydroxylase activity Geley et al. (1996)
Arg374Gln Lebanese Arab, female 6 K-helix Complete loss of 11␤-hydroxylase activity Curnow et al. (1993)
Arg384Gln Adult female 7 Heme propionate neutralisation Complete loss of 11␤-hydroxylase activity Curnow et al. (1993)
Val441Glu Adult male 8 Meander Complete loss of 11␤-hydroxylase activity Curnow et al. (1993)
Arg448Cys Iranian, male 8 Heme propionate neutralisation Complete loss of 11␤-hydroxylase activity Geley et al. (1996)
Arg448His Moroccan Jew 8 Heme propionate neutralisation Complete loss of 11␤-hydroxylase activity White et al. (1991)
The mutations were found in patients suffering from CAH and were subsequently demonstrated to lead to abolished 11␤-hydroxylase activity in cell
culture experiments.

patients suffering from genetic aberrations in the CYP11B1 et al., 1997). Amino acids V129 and A331 are highly con-
and CYP11B2 genes. served among members of the CYP11B family, but not
11␤-Hydroxylase deficiency, besides 21-hydroxylase de- between CYP11B1 and other cytochrome P450 enzymes.
ficiency, is the second most common cause of CAH, an in- This is an indirect indication of the functional importance
herited disease with the inability to synthesise cortisol from of these amino acids, but the precise reason for their dis-
11-deoxycorticosterone. Characteristically this disease leads ruptive effects remains to be elucidated. Amino acid N133
to androgen excess and hypertension. Besides the mutations is also conserved among members of the CYP11B family
mentioned above, many other missense mutations which and seems to lie within the region that may form part of
have been found in patients suffering from this disease, lead the substrate access channel (Joehrer et al., 1997). The con-
to functionally disturbed enzymes after expression in cell served homologous residue to T318 in human CYP11B1 is
culture (see Table 1). Curnow et al. (1993) demonstrated that T252 in CYP101 and this residue has been shown to be in-
mutations T318M, R374Q, R384Q and V441G in exons 5, volved in proton transfer which is necessary for enzymatic
6, 7 and 8 lead to complete dysfunctional CYP11B1 activity. activity (Raag et al., 1991; Ravichandran et al., 1993). The
Mutations V129M, A331V, E371G and R448C were shown effect of mutations E371 and R374 in the K-helix may be
by Geley et al. (1996) to be defective for 11␤-hydroxylase well rationalised by the fact that these residues, highly con-
activity. Mutations P42S, N133H and T319M lead only to served among most cytochrome P450 families form part of
partially disturbed 11␤-hydroxylase activity (Joehrer et al., the ERR-triad and are thought to play a fundamental role
1997). As can be seen in Table 1, mutations leading to in enzyme stabilisation (Hasemann et al., 1995).
11␤-hydroxylase deficiency are distributed over the en- Isolated deficiencies of aldosterone biosynthesis caused
tire coding region, but with a slightly enhanced frequency by CYP11B2 gene defects (see Table 2) are called aldos-
in exons 6 and 8. This might reflect the presence of terone synthase deficiencies type I and type II, formerly
functionally important amino acid residues in these re- called corticosterone methyl oxidase (CMO) deficiencies,
gions or, alternatively, mutations in this region are more which lead to hypoaldosteronism. Since the nomenclature
likely to have deleterious effects on enzymatic activity. and classification of these different diseases is being dis-
Amino acid residue P42 in the A-helix is speculated to cussed controversially (Portrat-Doyen et al., 1998), they
be essential for proper membrane orientation (Joehrer will be herein referred to only as aldosterone synthase

Table 2
List of published missense mutations in the human CYP11B2 gene
Mutation Patient Exon Putative 3D location Effect in vitro Reference

Thr185Ile Boy 3 E-helix Reduced 18-hydroxylase activity, no detectable Peter et al. (1998), Hampf
18-oxidase activity et al., unpublished results
Arg384Pro Caucasian, male 7 ␤1-4 Complete loss of aldosterone synthase activity Geley et al. (1995)
Leu461Pro Turk 8 L-helix Complete loss of aldosterone synthase activity Nomoto et al. (1997)
Val386Ala/Arg181Trp Iranian Jew 7/3 ␤1-4/between Increased 11␤- and 18-hydroxylase activity, no Pascoe et al. (1992a)
D- and E-helix detectable 18-oxidase activity
Val386Ala/Glu198Asp French twin boy 7/3 ␤1-4/E-helix Increased 11␤- and 18-hydroxylase activity, no Portrat-Doyen et al. (1998)
detectable 18-oxidase activity
The mutations were found in patients suffering from aldosterone synthase deficiency and were subsequently demonstrated to lead to abolished aldosterone
synthase activity in cell culture experiments.
154 M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159

deficiencies. These disorders lead to decreased sodium re-


sorption and potassium secretion to the urine. The plasma
renin activity is elevated and the plasma levels of aldos-
terone precursors are elevated. All patients suffered in
infancy from failure to thrive. The mutation Thr185Ile has
been found in a boy suffering from aldosterone synthase
deficiency (Peter et al., 1998), and subsequent investiga-
tions in our laboratory showed that this mutation leads to
a severe reduction of 18-hydroxylation and to a complete
loss of aldosterone synthase activity (Hampf et al., un-
published results). Although CYP11B1 and CYP11B2 are
invariant at this position, the mutated CYP11B2 enzyme
displayed much stronger 11␤-hydroxylase activity than
wild type CYP11B2 towards both 11-deoxycorticosterone
and 11-deoxycortisol. The mutation Arg384Pro leads to al-
dosterone deficiency (Geley et al., 1995). Double mutations Fig. 5. Presentation of relative aldosterone synthase activities of wild
type CYP11B2 and effect of the mutants Ile112Pro, Asp147Glu and
Glu198Asp and Val386Ala lead to enzymes with decreased Ile112Pro/Asp147Glu on 11␤-hydroxylation, 18-hydroxylation and 18-ox-
11␤- and 18-hydroxylase activity and impaired 18-oxidase idation towards 11-deoxycorticosterone (DOC) expressed in COS-1 cells.
activity (Portrat-Doyen et al., 1998), however, none of
these mutations alone abolished aldosterone synthase ac-
tivity completely. Mutations Arg181Trp and Val386Ala aldosterone synthesis may be modified independently, af-
were found in Iranian Jews suffering from aldosterone fecting only one reaction step, or differentially by altering
deficiency. Subsequent analysis of the CYP11B2 genes the three hydroxylation steps to distinct degrees.
in COS-1 cells showed that Arg181Trp does not affect The effect of the mutation Lys251Arg in CYP11B2
11␤-hydroxylation activity, but reduces 18-hydroxylation was investigated by Fardella et al. (1995). It caused four
and 18-oxidation activity. Additional presence of Val386Ala times as much 18-hydroxycorticosterone formation and
abolishes 18-oxidation activity completely (Pascoe et al., 50–80% increased aldosterone formation. If such a muta-
1992a). Taken together, it can be quoted that aldosterone tion would occur in patients, the predicted phenotype would
synthase deficiencies due to missense mutations are less be low-renin hypertension, due to the overproduction of
frequent than 11␤-hydroxylation deficiencies and that the aldosterone caused by this CYP11B2 mutant.
role of these highly conserved residues remains to be elu- Very interesting activity swapping mutants have been
cidated. A variety of mutations found in patients that suffer obtained by mutating amino acids located in the putative
from 11␤-hydroxylase and aldosterone synthase deficien- I-helix. It was found that single mutations at positions 296,
cies, like point mutations, deletions and incorrect splicing in 301, 302, 320, and 335 in CYP11B2 exhibited only slightly
the CYP11B1 and CYP11B2 genes leading to completely increased 11␤-hydroxylase activities in CYP11B2 when
inactive enzymes are summarised by Peter et al. (1999). exchanged to the corresponding ones found in CYP11B1
In an effort to identify the minimum number of mutations (Böttner et al., 1996). However, a Leu301Pro/Ala320Val
relevant for the different specific hydroxylation reactions be- double substitution increased 11␤-hydroxylase activity
tween the CYP11B-family members, residue-swapping mu- to 60% as compared to that of wild type CYP11B1,
tants between these isoenzymes have been investigated us- and the additional substitution Glu302Asp further en-
ing site-directed mutagenesis. hanced this activity to 85%. Concomitantly, the aldos-
Residues differing between human CYP11B1 and terone synthase activities of the mutant CYP11B2 pro-
CYP11B2 localised in the N-terminal part of the enzymes teins were suppressed to varying degrees, with double
have been investigated by Bechtel et al. (2002) by transient and triple replacement mutants Leu301Pro/Ala320Val and
expression of CYP11B2 mutants in COS-1 cells. The study Leu301Pro/Glu302Asp/Ala320Val retaining only about
revealed a three-fold increased 11␤-hydroxylation after 10% of CYP11B2 wild type activity (Fig. 6). Thus, a
substitution of Ile112 by proline and an even a four-fold in- mineralocorticoid-synthesising P450 has been engineered to
creased activity is exhibited by mutant Asp147Glu. A com- a glucocorticoid-synthesising enzyme by only two to three
bination of both mutations increased the 11␤-hydroxylation point mutations (Böttner et al., 1996). In reciprocal residue
activity towards 11-deoxycorticosterone six-fold. The ef- swapping experiments, CYP11B1 mutant Val320Ala, either
fect on the second and third catalytic step of CYP11B2 alone (Böttner and Bernhardt, 1996) (Fig. 6) or in com-
was less pronounced, except for Ile112Pro which showed bination with mutation Ser288Gly (Curnow et al., 1997;
an about two-fold increased 18-hydroxylation activity Mulatero et al., 1998) or Asn335Asp (Böttner et al., 1998),
(Fig. 5). Replacements Ile112Ser and Lys152Asn did not conferred aldosterone synthase activity to CYP11B1, para-
alter CYP11B2 substrate conversion. These investigations doxically without significantly affecting 11␤-hydroxylase
demonstrated that the three enzymatic reaction steps of activity. Thus, a glucocorticoid-synthesising enzyme has
M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159 155

is essential in conferring aldosterone synthase activity to


CYP11B1.

5.2. Activity modulation of CYP11B1 and CYP11B2 by


regulation of the electron transfer rate

As mentioned above, mitochondrial cytochrome P450-


dependent steroid hydroxylases require a NADPH-dependent
redox system consisting of adrenodoxin reductase and
adrenodoxin (Adx) (Bernhardt, 1996). It has been shown
that positively charged residues on the proximal surface
of the cytochrome P450 on the one hand (Wada and
Waterman, 1992; Lepesheva et al., 2000; Azeva et al.,
2001) and both negatively charged amino acids (Coghlan
and Vickery, 1991, 1992) as well as hydrophobic residues
on the surface of Adx on the other hand (Beckert et al.,
1994; Beckert and Bernhardt, 1997; Bernhardt, 1998) are
important for the correct function of the ferredoxin as an
efficient electron carrier.
In COS-1 cells the source of reducing equivalents is
limited. The activity of the bovine and human CYP11B
isoenzymes is stimulated by an enhanced electron trans-
fer due to an increased expression of cotransfected Adx
(Zuber et al., 1988; Mathew et al., 1990). In addition,
in case of bovine CYP11B1 the ratio of corticosterone
to 18-hydroxycorticosterone and to aldosterone decreases
with a high level of cotransfected Adx. This is also the
case for human CYP11B2. Cotransfected Adx stimulated
18-hydroxylation and 18-oxidation activity besides in-
creasing 11␤-hydroxylation activity. This indicates that the
availability of a sufficient amount of electrons supports
the formation of 18-hydroxycorticosterone and aldosterone
(Cao and Bernhardt, 1999b).
Fig. 6. Presentation of aldosterone and cortisol formation of wild The effects caused by truncated forms of Adx (e.g. Adx
type human CYP11B2 and CYP11B1. CYP11B2, the mineralocorti- 4-108 and Adx 4-114) have been investigated with respect
coid synthesising protein has been changed to a glucocorticoid syn- to their reduction rate of CYP11B1. The truncated mutants
thesising one by two or three point mutations (Leu301Pro/Ala320Val Adx 4-108 and Adx 4-114 led to an increase in corti-
and Leu301Pro/Glu302Asp/Ala320Val). CYP11B1 became an aldosterone
synthase by a single point mutation (Val320Ala).
costerone formation from 11-deoxycorticosterone using a
liposomal reconstitution assay with bovine CYP11B1 (Cao
and Bernhardt, 1999a). The apparent Vmax values of al-
been engineered to a mineralocorticoid-synthesising en- dosterone formation by bovine CYP11B1 were determined
zyme by a single point mutation. to be three-fold higher for Adx 4-108 and two-fold higher
Diverging residues in the C-terminal regions of CYP11B1 for Adx 4-114 as compared to wild type Adx. Subsequent
and CYP11B2 at positions 471, 472, 492, 493 and 494, investigations in COS-1 cells confirmed the stimulating
have been investigated, but were shown to be insignificant effect of cotransfected Adx 1-108 on 11␤-hydroxylation
for the stereo- and regiospecificity of steroid hydroxyla- activity of bovine CYP11B1, human CYP11B1 and human
tion (Böttner et al., 1998). These investigations, in combi- CYP11B2 as well as an enhancement of 18-hydroxylation
nation with naturally occurring chimeras of CYP11B1 and and 18-oxidation activity of bovine CYP11B1 and human
CYP11B2, suggested that there are residues spread through- CYP11B2 as compared to cotransfection with wild type Adx
out the protein which are important for proper stereoselec- (Cao and Bernhardt, 1999a). Both 18-hydroxycorticosterone
tive hydroxylation activity. However, the residue swapping and aldosterone are correlated with hypertension. Several
experiments suggest that side chains located in the puta- C-terminally truncated species of Adx, e.g. species with 114,
tive I-helix of the CYP11B enzymes contribute most signif- 121, 124, 125 and 127 amino acid residues, respectively,
icantly to substrate recognition and conversion. The I-helix have been isolated from bovine adrenal cortex (Tanaka et al.,
is translated from exon 5 and exon 6 which further un- 1973; Hiwatashi et al., 1986; Sakihama et al., 1988). Un-
derlines the findings of Pascoe et al. (1992b) that exon 5 der circumstances where Adx is progressively C-terminally
156 M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159

truncated in vivo the accompanying increased electron be differentially regulated on the level of membrane compo-
transfer ability of the truncated Adx could lead to regulatory sition: in a phospholipid vesicle system, a strong dependence
relevance in a way that the respective adrenal glands would of aldosterone synthesis on the composition of the lipids and
secrete more aldosterone (Uhlmann et al., 1994). However, the presence of calmodulin was observed (Ohnishi et al.,
it is not known whether the truncated species of Adx are 1984, 1986; Wada et al., 1984; Seybert, 1990). Another hy-
artefacts or whether physiological conditions leading to a pothesis suggests that CYP11B2 activity may be controlled
changed electron flow in adrenal tissue do occur. post-translationally by protein phosphorylation producing
Taken togeher, the results obtained with different Adx short-term activity control (Bureik et al., 2002b). Further
mutants, which display different electron transfer properties studies are, however, necessary to support this finding.
support the proposal that an increase in the electron transfer
rate leads to an enhancement of the ‘intermediate flow’ to-
wards aldosterone. This observation could be of physiologi- 6. Outlook
cal importance since in bovine adrenal cortex the molar ratio
of AdR/Adx/CYP11B1 is approximately 1:3:4 (Hanukoglu Overexpression of CYP11B1 and CYP11B2 in bacteria
and Hanukoglu, 1986). The corresponding ratio in human has not been successful so far. Only expression of the rat
tissue has not been determined. An increase of the concen- homologues was achieved at low yield in Escherichia coli
tration of wild type Adx, e.g. in the case of ACTH-dependent (Nonaka et al., 1998). Besides in mammalian cells the func-
induction, could exert a significant effect on aldosterone pro- tional expression of human CYP11B1 and CYP11B2 using
duction under these conditions. the fission yeast Schizosaccharomyces pombe (Bureik et al.,
2002c) was demonstrated. It was found that the transformed
5.3. Activity modulation by protein–protein interaction yeast show strong steroid hydroxylase activity in vivo in
with CYP11A1 comparison to transfected mammalian cells (see above). So
far, most studies performed with these enzymes used recom-
The aspect of activity modulation of CYP11B enzymes binant cell cultures, and therefore detailed studies on the
by interaction with CYP11A1, the cholesterol side chain biochemical reaction mechanism and properties (e.g. sub-
cleavage enzyme, which is the only other mitochondrial strate binding, protein–protein interactions, electron trans-
P450 in the adrenal cortex, has been studied intensively. fer, substrate induced structural changes and EPR spectra)
Competition between rat CYP11A1 and CYP11B1 for re- are not available. It can be expected that development in
ducing equivalents is observed in vitro (Yamazaki et al., this field will allow the production and isolation of human
1993). Additionally, transfection with the human or bovine CYP11Bs and their mutants and improve our knowledge on
enzymes in COS-1 cells containing a limited amount of the structure–function relationship of these highly homolo-
reducing equivalents resulted in a decrease of cortisol and gous but nevertheless diverse enzymes.
aldosterone synthesis, respectively (Cao and Bernhardt, As shown above, different mechanisms may lead to over-
1999b). In contrast to this, the bovine enzymes displayed production of aldosterone. A pathologically high level of
a specific interaction indicating allosteric effects of bovine plasma aldosterone, on the other hand, may not only lead to
CYP11A1 on bovine CYP11B1 in reconstituted liposo- hypertension, but could also cause progression of congestive
mal membranes (Ikushiro et al., 1992; Kominami et al., heart failure and myocardial fibrosis, as demonstrated re-
1994) and in COS-1 cells capable of high electron transfer cently. Treatment with antagonists of the mineralocorticoid
rates due to cotransfection of Adx (Cao and Bernhardt, receptor leads to decreased morbidity and mortality of the
1999b). In the liposomal membranes and in COS-1 cells patients (Pitt, 2002), but is accompanied by severe side ef-
bovine CYP11A1 stimulated 11␤-hydroxylation activity of fects. Thus, inhibition of aldosterone production by inhibit-
bovine CYP11B1, and decreased 18-hydroxycorticosterone ing CYP11B2 might be a more sophisticated approach to
and aldosterone formation at the same time. Therefore, it tackle this problem. Initial studies using recombinant V79
is tempting to speculate that CYP11A1 induces confor- and S. pombe cells showed that it is possible to find selec-
mational changes in CYP11B1 which differentially affect tive CYP11B inhibitors (Denner et al., 1995; Denner and
binding of the substrate and the intermediates, so that Bernhardt, 1998; Ehmer et al., 2002), which could interfere
corticosterone can leave the active site in favour of being with the fatal processes of mineralocorticoid receptor an-
further 18-hydroxylated and 18-oxidised. However, human tagonists. An overview about the recent development of in-
CYP11A1 does not show such a specific interaction with hibitors of both, CYP11B1 and CYP11B2, is given in Bureik
human CYP11B2. There is only competition for reducing et al. (2002a).
equivalents at low concentrations of electrons supplied and
an increase in all enzymatic activities at high electron sup-
ply, but no effect on the product pattern (Cao and Bernhardt, Acknowledgements
1999b).
Modulation of CYP11B2 activity has also been shown to We thank Matthias Bureik and Andy Zöllner for critical
depend on other circumstances. Aldosterone formation may reading of the manuscript.
M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159 157

References Cao, P.-R., Bernhardt, R., 1999b. Interaction of CYP11B1 (cytochrome


P-45011 b) with CYP11A1 (cytochrome P-450scc) in COS-1 cells.
Eur. J. Biochem. 262, 720–726.
Akhrem, A.A., Lapko, V.N., Lapko, A.G., Shkumatov, V.M., Chashchin,
Coghlan, V.M., Vickery, L.E., 1991. Site-specific mutations in human
V.L., 1979. Isolation, structural organization and mechanism of action
ferredoxin that affect binding to ferredoxin reductase and cytochrome
of mitochondrial steroid hydroxylating systems. Acta Biol. Med. Ger.
P450scc. J. Biol. Chem. 266, 18606–18612.
38, 257–273.
Coghlan, V.M., Vickery, L.E., 1992. Electrostatic interactions stabilizing
Azeva, T.N., Gilep, A.A., Lepesheva, G.I., Strushkevich, N.V., Usanov,
ferredoxin electron transfer complexes. Disruption by ‘conservative’
S.A., 2001. Site-directed mutagenesis of cytochrome P450scc. II. Ef-
mutations. J. Biol. Chem. 267, 8932–8935.
fect of replacement of the Arg425 and Arg426 residues on the struc-
Connell, J.M., Fraser, R., MacKenzie, S., Davies, E., 2003. Is altered
tural and functional properties of the cytochrome P450scc. Biochem-
adrenal steroid biosynthesis a key intermediate phenotype in hyper-
istry (Moscow) 66, 546–575.
tension? Hypertension 41, 993–999.
Bechtel, S., Belkina, N., Bernhardt, R., 2002. The effect of amino acid
Cosme, J., Johnson, E.F., 2000. Engineering microsomal cytochrome P450
substitutions I112P, D147E and K152N in CYP11B2 on the catalytic
2C5 to be a soluble, monomeric enzyme. Mutations that alter aggre-
activities of the enzyme. Eur. J. Biochem. 269, 1118–1127.
gation, phospholipid dependence of catalysis, and membrane binding.
Beckert, V., Dettmer, R., Bernhardt, R., 1994. Mutations of tyrosine 82 in J. Biol. Chem. 275, 2545–2553.
bovine adrenodoxin that affect binding to cytochromes P45011A1 and
Curnow, K.M., Tusie-Luna, M.T., Pascoe, L., Natarajan, R., Gu, J.L.,
P45011B1 but not electron transfer. J. Biol. Chem. 269, 2568–2573.
Nadler, J.L., White, P.C., 1991. The product of the CYP11B2 gene
Beckert, V., Bernhardt, R., 1997. Specific aspects of electron transfer from is required for aldosterone biosynthesis in the human adrenal cortex.
adrenodoxin to cytochromes p450scc and p45011beta. J. Biol. Chem. Mol. Endocrinol. 5, 1513–1522.
272, 4883–4888. Curnow, K.M., Slutsker, L., Vitek, J., Cole, T., Speiser, P.W., New, M.I.,
Belkina, N.V., Lisurek, M., Ivanov, A.S., Bernhardt, R., 2001. Modelling White, P.C., Pascoe, L., 1993. Mutations in the CYP11B1 gene causing
of three-dimensional structures of cytochromes P450 11B1 and 11B2. congenital adrenal hyperplasia and hypertension cluster in exons 6, 7,
J. Inorg. Biochem. 87, 197–207. and 8. Proc. Natl. Acad. Sci. U.S.A. 90, 4552–4556.
Bernhardt, R., 1996. Cytochrome P450: structure, function, and generation Curnow, K.M., Mulatero, P., Emeric-Blanchouin, N., Aupetit-Faisant, B.,
of reactive oxygen species. Rev. Physiol. Biochem. Pharmacol. 127, Corvol, P., Pascoe, L., 1997. The amino acid substitutions Ser288Gly
137–221. and Val320Ala convert the cortisol producing enzyme, CYP11B1, into
Bernhardt, R., 1998. Electron transfer in mitochondrial steroid hydroxy- an aldosterone producing enzyme. Nat. Struct. Biol. 4, 32–35.
lating cytochrome P450 systems: role of adrenodoxin. In: Nicolini, C. Delles, C., Erdmann, J., Jacobi, J., Hilgers, K.F., Fleck, E.,
(Ed.), Biophysics of Electron Transfer and Molecular Bioelectronics. Regitz-Zagrosek, V., Schmieder, R.E., 2001. Aldosterone synthase
Plenum Press, New York, pp. 51–66. (CYP11B2) -344 C/T polymorphism is associated with left ventricu-
Boon, W.C., Roche, P.J., Hammond, V.E., Jeyaseelan, K., Crawford, R.J., lar structure in human arterial hypertension. J. Am. Coll. Cardiol. 37,
Coghlan, J.P., 1995. Cloning and expression analysis of a cytochrome 878–884.
P-450(11 beta) cDNA in sheep. Biochim. Biophys. Acta 1260, 109– Denner, K., Doehmer, J., Bernhardt, R., 1995. Cloning of CYP11B1 and
112. CYP11B2 from normal human adrenal and their functional expression
Böttner, B., Schrauber, H., Bernhardt, R., 1996. Engineering a in COS-7 and V79 Chinese hamster cells. Endocr. Res. 21, 443–448.
mineralocorticoid- to a glucocorticoid-synthesizing cytochrome P450. Denner, K., Bernhardt, R., 1998. Inhibition studies of steroid conversions
J. Biol. Chem. 271, 8028–8033. mediated by human CYP11B1 and CYP11B2 expressed in cell cul-
Böttner, B., Bernhardt, R., 1996. Changed ratios of glucocorti- tures. In: Ishimura, Y., Shimada, H., Suematsu, M. (Eds.), Oxygen
coid/mineralocorticoids caused by point mutations in the putative Homeostasis and Its Dynamics. Springer-Verlag, Tokyo, Berlin, Hei-
I-helix regions of CYP11B1 and CYP11B2. Endocr. Res. 22, 455–561. delberg, New York, pp. 231–236.
Böttner, B., Denner, K., Bernhardt, R., 1998. Conferring aldosterone Domalik, L.J., Chaplin, D.D., Kirkman, M.S., Wu, R.C., Liu, W.W.,
synthesis to human CYP11B1 by replacing key amino acid residues Howard, T.A., Seldin, M.F., Parker, K.L., 1991. Different isozymes
with CYP11B2-specific ones. Eur. J. Biochem. 252, 458–466. of mouse 11␤-hydroxylase produce mineralocorticoids and glucocor-
Brilla, C.G., 2000. Renin–angiotensin–aldosterone system and myocardial ticoids. Mol. Endocrinol. 5, 1853–1861.
fibrosis. Cardiovasc. Res. 47, 1–3. Ehmer, P.B., Bureik, M., Bernhardt, R., Müller, U., Hartmann, R.W., 2002.
Bülow, H.E., Mobius, K., Bahr, V., Bernhardt, R., 1996. Molecular cloning Development of a test system for inhibitors of human aldosterone
and functional expression of the cytochrome P450 11␤-hydroxylase synthase (CYP11B2): screening in fission yeast and evaluation of
of the guinea pig. Biochem. Biophys. Res. Commun. 221, 304–312. selectivity in V79 cells. J. Steroid Biochem. Mol. Biol. 81, 173–179.
Bülow, H.E., Bernhardt, R., 2002. Analysis of the CYP11B gene family Erdmann, B., Denner, K., Gerst, H., Lenz, D., Bernhardt, R., 1995a.
in the guinea pig suggest the existence of a primordial CYP11B gene Human adrenal CYP11B1: localization by in situ-hybridization and
with aldosterone synthase activity. Eur. J. Biochem. 269, 3838–3846. functional expression in cell cultures. Endocr. Res. 21, 425–435.
Bureik, M., Lisurek, M., Bernhardt, R., 2002a. The human steroid hy- Erdmann, B., Gerst, H., Bülow, H., Lenz, D., Bahr, V., Bernhardt, R.,
droxylases CYP11B1 and CYP11B2. Biol. Chem. 383, 1537–1551. 1995b. Zone-specific localization of cytochrome P45011B1 in human
Bureik, M., Zeeh, A., Bernhardt, R., 2002b. Modulation of steroid hydrox- adrenal tissue by PCR-derived riboprobes. Histochem. Cell. Biol. 104,
ylase activity in stably transfected V79MZh11B1 and V79MZh11B2 301–307.
cells by PKC and PKD inhibitors. Endocr. Res. 28, 351–355. Erdmann, B., Gerst, H., Lippoldt, A., Bülow, H., Ganten, D., Fuxe, K.,
Bureik, M., Schiffler, B., Hiraoka, Y., Vogel, F., Bernhardt, R., 2002c. Bernhardt, R., 1996. Expression of cytochrome P45011B1 in the brain
Functional expression of human mitochondrial CYP11B2 in fission of normal hypertensive transgenic rats. Brain Res. 733, 73–82.
yeast and identification of a new internal electron transfer protein, Fardella, C.E., Rodriguez, H., Hum, D.W., Mellon, S.H., Miller, W.L.,
etp1. Biochemistry 41, 2311–2321. 1995. Artificial mutations in P450AS (aldosterone synthase) can in-
Chua, S.C., Szabo, P., Vitek, A., Grzeschik, K.H., John, M., White, P.C., crease enzymatic activity: a model for low-renin hypertension? J. Clin.
1987. Cloning of cDNA encoding steroid 11␤-hydroxylase (P450c11). Endocrinol. Metab. 80, 1040–1043.
Proc. Natl. Acad. Sci. U.S.A. 84, 7193–7197. Ferrari, P., 2002. Genetics of the mineralocorticoid system in primary
Cao, P.-R., Bernhardt, R., 1999a. Modulation of aldosterone biosynthesis hypertension. Curr. Hypertens. Rep. 4, 18–24.
by adrenodoxin mutants with different electron transport efficiencies. Fisher, A., Friel, E.C., Bernhardt, R., Gomez-Sanchez, C., Connell, J.M.,
Eur. J. Biochem. 265, 152–159. Fraser, R., Davies, E., 2001. Effects of 18-hydroxylated steroids
158 M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159

on corticosteroid production by human aldosterone synthase and thase gene causes glucocorticoid-remediable aldosteronism and human
11␤-hydroxylase. J. Clin. Endocrinol. Metab. 86, 4326–4329. hypertension. Nature 355, 262–265.
Geley, S., Johrer, K., Peter, M., Denner, K., Bernhardt, R., Sippell, W.G., Lifton, R.P., Gharavi, A.G., Geller, D.S., 2001. Molecular mechanisms of
Kofler, R., 1995. Amino acid substitution R384P in aldosterone syn- human hypertension. Cell 104, 545–556.
thase causes corticosterone methyloxidase type I deficiency. J. Clin. Lijnen, P., Petrov, V., 2000. Induction of cardiac fibrosis by aldosterone.
Endocrinol. Metab. 80, 424–429. J. Mol. Cell. Cardiol. 32, 865–879.
Geley, S., Kapelari, K., Johrer, K., Peter, M., Glatzl, J., Vierhapper, H., Lim, P.O., Macdonald, T.M., Holloway, C., Friel, E., Anderson, N.H.,
Schwarz, S., Helmberg, A., Sippell, W.G., White, P.C., Kofler, R., Dow, E., Jung, R.T., Davies, E., Fraser, R., Connell, J.M., 2002.
1996. CYP11B1 mutations causing congenital adrenal hyperplasia due Variation at the aldosterone synthase (CYP11B2) locus contributes to
to 11beta-hydroxylase deficiency. J. Clin. Metab. 81, 2896–2901. hypertension in subjects with a raised aldosterone-to-renin ratio. J.
Gotoh, O., 1992. Substrate recognition sites in cytochrome P450 family Clin. Endocrinol. Metab. 87, 4398–4402.
2 (CYP2) proteins inferred from comparative analyses of amino acid MacKenzie, S.M., Clark, C.J., Fraser, R., Gomez-Sanchez, C.E., Con-
and coding nucleotide sequences. J. Biol. Chem. 267, 83–90. nell, J.M., Davies, E., 2000. Expression of 11beta-hydroxylase and
Hampf, M., Swart, A.C., Swart, P., 1996. Sequence of the aldosterone synthase genes in the rat brain. J. Mol. Endocrinol. 24,
11beta-hydroxylase gene from the cape baboon (Papio ursinus). En- 321–328.
docr. Res. 22, 495–499. MacConnachie, A.A., Kelly, K.F., Mcnamara, A., Loughlin, S., Gates,
Hampf, M., Dao, N.T., Hoan, N.T., Bernhardt, R., 2001. Unequal L.J., Inglis, G.C., Jamieson, A., Connell, J.M., Haites, N.E., 1998.
crossing-over between aldosterone synthase and 11beta-hydroxylase Rapid diagnosis and identification of cross-over sites in patients with
genes causes congenital adrenal hyperplasia. J. Clin. Endocrinol. glucocorticoid remediable aldosteronism. J. Clin. Endocrinol. Metab.
Metab. 86, 4445–4452. 83, 4328–4331.
Hanukoglu, I., Hanukoglu, Z., 1986. Stoichiometry of mitochondrial cy- Mathew, P.A., Mason, J.I., Trant, J.M., Sanders, D., Waterman, M.R., 1990.
tochromes P-450, adrenodoxin and adrenodoxin reductase in adrenal Amino acid substitutions Phe66Leu and Ser126Pro abolish cortisol
cortex and corpus luteum. Implications for membrane organization and aldosterone synthesis by bovine cytochrome P450(11)b. J. Biol.
and gene regulation. Eur. J. Biochem. 157, 27–31. Chem. 265, 20228–20233.
Hasemann, C.A., Kurumbail, R.G., Boddupalli, S.S., Peterson, J.A., Matsukawa, N., Nonaka, Y., Ying, Z., Higaki, J., Ogihara, T., Okamoto,
Deisenhofer, J., 1995. Structure and function of cytochromes P450: a M., 1990. Molecular cloning and expression of cDNAS encoding rat
comparative analysis of three crystal structures. Structure 3, 41–62. aldosterone synthase: variants of cytochrome P-450(11 b). Biochem.
Hautanen, A., Toivanen, P., Manttari, M., Tenkanen, L., Kupari, M., Biophys. Res. Commun. 169, 245–252.
Manninen, V., Kayes, K.M., Rosenfeld, S., White, P.C., 1999. Joint Mellon, S.H., Bair, S.R., Monis, H., 1995. P450c11B3 mRNA, transcribed
effects of an aldosterone synthase (CYP11B2) gene polymorphism from a third P450c11 gene, is expressed in a tissue-specific, devel-
and classic risk factors on risk of myocardial infarction. Circulation opmentally, and hormonally regulated fashion in the rodent adrenal
100, 2213–2218. and encodes a protein with both 11-hydroxylase and 18-hydroxylase
Hiwatashi, A., Sakihama, N., Shin, M., Ichikawa, Y., 1986. Heterogeneity activities. J. Biol. Chem. 270, 1643–1649.
of adrenocortical ferredoxin. FEBS Lett. 209, 311–315. Miller, W.L., Tyrell, J.B., 1995. The adrenal cortex. In: Felig, P., Bax-
Imai, T., Yamazaki, T., Kominami, S., 1998. Kinetic studies on ter, J.D., Frohmann, L.A., Broadus, A.E. (Eds.), Endocrinology and
bovine cytochrome p45011 b catalyzing successive reactions from Metabolism, third ed. McGraw-Hill Press, New York, USA, pp.
11-deoxycorticosterone to aldosterone. Biochemistry 37, 8097–8104. 642–659.
Ikushiro, S., Kominami, S., Takemori, S., 1992. Adrenal P-450scc modu- Mornet, E., Dupont, J., Vitek, A., White, P.C., 1989. Characterization of
lates activity of P-45011 b in liposomal and mitochondrial membranes. two genes encoding human steroid 11 ␤-hydroxylase [P-450(11) ␤].
Implication of P-450scc in zone specificity of aldosterone biosynthesis J. Biol. Chem. 264, 20961–20967.
in bovine adrenal. J. Biol. Chem. 267, 1464–1469. Mulatero, P., Curnow, K.M., Aupetit-Taisant, B., Foekling, M., 1998.
Joehrer, K., Geley, S., Strasser-Wozak, E.M., Azziz, R., Wollmann, H.A., Recombinant CYP11B genes encode enzymes that can catalyze
Schmitt, K., Kofler, R., White, P.C., 1997. CYP11B1 mutations causing conversion of 11-deoxycortisol to cortisol, 18-hydroxycortisol, and
non-classic adrenal hyperplasia due to 11beta-hydroxylase deficiency. 18-oxocortisol. J. Clin. Endocinol. Metab. 83, 3996–4001.
Hum. Mol. Genet. 6, 1829–1834. Nakagawa, Y., Yamada, M., Ogawa, H., Igarashi, Y., 1995. Missense
Kawamoto, T., Mitsuuchi, Y., Ohnishi, T., Ichikawa, Y., Yokoyama, Y., mutation in CYP11B1 (CGA[Arg384] GCA[Gly]) causes steroid
Sumimoto, H., Toda, K., Miyahara, K., Kuribayashi, I., Nakao, K., 11beta-hydroxylase deficiency. Eur. J. Endocrinol. 132, 286–289.
1990a. Cloning and expression of a cDNA for human cytochrome Nomoto, S., Massa, G., Mitani, F., Ishimura, Y., Miyahara, K., Toda,
P-450aldo as related to primary aldosteronism. Biochem. Biophys. K., Nagano, I., Yamashiro, T., Ogoshi, S., Fukata, J., Onishi, S.,
Res. Commun. 173, 309–316. Hashimoto, K., Doi, Y., Imura, H., Shizuta, Y., 1997. CMO I deficiency
Kawamoto, T., Mitsuuchi, Y., Toda, K., Miyahara, K., Yokoyama, Y., caused by a point mutation in exon 8 of the human CYP11B2 gene
Nakao, K., Hosoda, K., Yamamoto, Y., Imura, H., Shizuta, Y., 1990b. encoding steroid 18-hydroxylase (P450C18). Biochem. Biophys. Res.
Cloning of cDNA and genomic DNA for human cytochrome P-45011 Commun. 234, 382–385.
␤. FEBS Lett. 269, 345–349. Nonaka, Y., Takemori, H., Halder, S.K., Sun, T., Ohta, M., Hatano,
Kominami, S., Harada, D., Takemori, S., 1994. Regulation mechanism O., Takakusu, A., Okamoto, M., 1995. Frog cytochrome P-450 (11
of the catalytic activity of bovine adrenal cytochrome P-450(11)b. b,aldo), a single enzyme involved in the final steps of glucocorticoid
Biochim. Biophys. Acta 1192, 234–240. and mineralocorticoid biosynthesis. Eur. J. Biochem. 229, 249–256.
Kupari, M., Hautanen, A., Lankinen, L., Koskinen, P., Virolainen, J., Nonaka, Y., Fujii, T., Kagawa, N., Waterman, M.R., Takemori, H.,
Nikkila, H., White, P.C., 1998. Associations between human aldos- Okamoto, M., 1998. Structure/function relationship of CYP11B1 as-
terone synthase (CYP11B2) gene polymorphisms and left ventricular sociated with Dahl’s salt-resistant rats—expression of rat CYP11B1
size, mass, and function. Circulation 97, 569–575. and CYP11B2 in Escherichia coli. Eur. J. Biochem. 258, 869–878.
Lepesheva, G.I., Azeva, T.N., Srushkevich, N.V., Gilep, A.A., Us- Ogishima, T., Shibata, H., Shimada, H., Mitani, F., Suzuki, H., Saruta, T.,
anov, S.A., 2000. Site-directed mutagenesis of cytochrome P450scc Ishimura, Y., 1991. Aldosterone synthase cytochrome P-450 expressed
(CYP11A1). Effect of lysine residue substitution on its structural and in the adrenals of patients with primary aldosteronism. J. Biol. Chem.
functional properties. Biochemistry (Moscow) 65, 1409–1418. 266, 10731–10734.
Lifton, R.P., Dluhy, R.G., Powers, M., Rich, G.M., Cook, S., Ulick, S., Ohnishi, T., Wada, A., Nonaka, Y., Okamoto, M., Yamano, T.,
Lalouel, J.M., 1992. A chimaeric 11␤-hydroxylase/aldosterone syn- 1984. Effect of phospholipid on aldosterone biosynthesis by a cy-
M. Lisurek, R. Bernhardt / Molecular and Cellular Endocrinology 215 (2004) 149–159 159

tochrome P- 450(11) ␤-reconstituted system. Biochem. Int. 9, 715– Tanaka, M., Haniu, M., Yasunobu, K.T., Kimura, T., 1973. The amino
723. acid sequence of bovine adrenodoxin. J. Biol. Chem. 248, 1141–
Ohnishi, T., Wada, A., Nonaka, Y., Sugiyama, T., Yamano, T., Okamoto, 1157.
M., 1986. Effect of calmodulin on aldosterone synthesis by a cy- Taymans, S.E., Pack, S., Pak, E., Torpy, D.J., Zhuang, Z., Stratakis, C.A.,
tochrome P-450(11) ␤-reconstituted system from bovine adrenocorti- 1998. Human CYP11B2 (aldosterone synthase) maps to chromosome
cal mitochondria. J. Biochem. (Tokio) 100, 1065–1076. 8q24.3. J. Clin. Endocrinol. Metab. 83, 1033–1036.
Ortiz De Montellano, P.R., 1995. Oxygen activation and reactivity. In: Uhlmann, H., Kraft, R., Bernhardt, R., 1994. C-terminal region of adren-
Ortiz De Montellano, P.R. (Ed.), Cytochrome P450: Stucture, Mecha- odoxin affects its structural integrity and determines differences in its
nism and Biochemistry. Plenum Press, New York, USA, pp. 245–303. electron transfer function to cytochrome P-450. J. Biol. Chem. 269,
Ozaki, H.S., Iwahashi, K., Tsubaki, M., Fukui, Y., Ichikawa, Y., Takeuchi, 22557–22564.
Y., 1991. Cytochrome P-45011 beta in rat brain. J. Neurosci. Res. 28, Veronneau, S., Bernard, H., Cloutier, M., Courtemanche, J., Ducharme, L.,
518–524. Lefebvre, A., Mason, J.I., LeHoux, J.G., 1996. The hamster adrenal cy-
Pascoe, L., Curnow, K.M., Slutsker, L., Connell, J.M., Rösler, A., White, tochrome P450C11 has equipotent 11-hydroxylase and 19-hydroxylase
P.C., 1992a. Mutations in the human CYP11B2 (aldosterone synthase) activities, but no aldosterone synthase activity. J. Steroid Biochem.
gene causing corticosterone methyloxidase II deficiency. Proc. Natl. Mol. Biol. 57, 125–139.
Acad. Sci. U.S.A. 89, 4996–5000. Wada, A., Okamoto, M., Nonaka, Y., Yamano, T., 1984. Aldosterone
Pascoe, L., Curnow, K.M., Slutsker, L., Connell, J.M., Speiser, P.W., biosynthesis by a reconstituted cytochrome P-45011 b system.
New, M.I., White, P.C., 1992b. Glucocorticoid-suppressible hyperal- Biochem. Biophys. Res. Commun. 119, 365–371.
dosteronism results from hybrid genes created by unequal crossovers Wada, A., Ohnishi, T., Nonaka, Y., Okamoto, M., Yamano, T., 1985.
between CYP11B1 and CYP11B2. Proc. Natl. Acad. Sci. U.S.A. 89, Synthesis of aldosterone by a reconstituted system of cytochrome
8327–8331. P- 45011 b from bovine adrenocortical mitochondria. J. Biochem.
Pascoe, L., Jeunemaitre, X., Lebrethon, M.C., Curnow, K.M., (Tokyo) 98, 245–256.
Gomez-Sanchez, C.E., Gasc, J.M., Saez, J.M., Corvol, P., 1995. Wada, A., Waterman, M.R., 1992. Identification by site-directed muta-
Glucocorticoid-suppressible hyperaldosteronism and adrenal tumours genesis of two lysine residues in cholesterol side chain cleavage cy-
occurring in a single French pedigree. J. Clin. Invest. 96, 2236–2246. tochrome P450 that are essential for adrenodoxin binding. J. Biol.
Peter, M., Bunger, K., Solyom, J., Sippell, W.G., 1998. Mutation Thr185I Chem. 267, 22877–22882.
is associated with corticosterone methyl oxidase deficiency type II. Wagner, M.J., Ge, Y., Siciliano, M., Wells, D.E., 1991. A hybrid cell map-
Eur. J. Pediatr. 157, 378–381. ping panel for regional localization of probes to human chromosome
Peter, M., Dubuis, J.-M., Sippell, W.G., 1999. Disorders of the aldosterone 8. Genomics 10, 114–125.
synthase and steroid 11␤-hydroxylase deficiencies. Horm. Res. 51, Waterman, M.R., Simson, E.R., 1989. Regulation of steroid hydroxylase
211–222. gene expression is multifunctional in nature. Recent Prog. Horm. Res.
Pitt, B., 2002. Do diuretics and aldosterone receptor antagonists improve 45, 533–563.
ventricular remodeling? J. Card. Fail. 8, 491–493. White, P.C., Dupont, J., New, M.I., Leibermann, E., Hochberg, Z., Rösler,
Portrat-Doyen, S., Tourniaire, T., Richard, O., Mulatero, P., A., 1991. A mutation in CYP11B1 (Arg448His) associated with steroid
Aupetit-Faisant, B., Curnow, K.M., Pascoe, L., Morel, Y., 1998. Iso- 11␤-hydroxylase deficiency in Jews of Moroccan origin. J. Clin. Invest.
lated aldosterone synthase deficiencies caused by simultaneous E198D 87, 1664–1667.
and V386A mutations in the CYP11B2 gene. J. Clin. Endocrinol. White, P.C., 2001. Steroid 11 ␤-hydroxylase deficiency and related dis-
Metab. 83, 4156–4161. orders. Endocrinol. Metab. Clin. North Am. 30, 61–79.
Quinn, S.J., Williams, G.H., 1988. Regulation of aldosterone secretion. Williams, P.A., Cosme, J., Sridhar, V., Johnson, E.F., McRee, D.E., 2000.
Annu. Rev. Physiol. 50, 409–426. Mammalian microsomal cytochrome P450 monooxygenase: structural
Raag, R., Martinis, S.A., Sligar, S.G., Poulos, T.L., 1991. Crystal struc- adaptations for membrane binding and functional diversity. Mol. Cell
ture of the cytochrome P-450CAM active site mutant Thr252Ala. 5, 121–131.
Biochemistry 30, 11420–11429. Williams, P.A., Cosme, J., Ward, A., Angove, H.C., Matak Vinkovic, D.,
Ravichandran, K.G., Boddupalli, S.S., Hasermann, C.A., Peterson, J.A., Jhoti, H., 2003. Crystal structure of human cytochrome P450 2C9
Deisenhofer, J., 1993. Crystal structure of hemoprotein domain of with bound warfarin. Nature 424, 464–468.
P450BM-3, a prototype for microsomal P450’s. Science 261, 731–736. Yamazaki, T., Mcnamara, B.C., Jefcoate, C.R., 1993. Competition for
Rudolph, A.E., Blasi, E.R., Delyani, J.A., 2000. Tissue-specific corticos- electron transfer between cytochromes P450scc and P45011 b in rat
teroidogenesis in the rat. Mol. Cell. Endocrinol. 165, 221–224. adrenal mitochondria. Mol. Cell. Endocrinol. 95, 1–11.
Sakihama, N., Hiwatashi, A., Miyatake, A., Shin, M., Ichikawa, Y., 1988. Yanagibashi, K., Haniu, M., Shively, J.E., Shen, W.H., Hall, P.,
Isolation and purification of mature bovine adrenocortical ferredoxin 1986. The synthesis of aldosterone by the adrenal cortex. Two
with an elongated carboxyl end. Arch. Biochem. Biophys. 264, 23–29. zones (fasciculata and glomerulosa) possess one enzyme for 11␤-,
Schunkert, H., Hengstenberg, C., Holmer, S.R., Broeckel, U., Luchner, 18-hydroxylation, and aldosterone synthesis. J. Biol. Chem. 261, 3556–
A., Muscholl, M.W., Kurzinger, S., Doring, A., Hense, H.W., Riegger, 3562.
G.A., 1999. Lack of association between a polymorphism of the Yoshimura, M., Nakamura, S., Ito, T., Nakayama, M., Harada, E., Mizuno,
aldosterone synthase gene and left ventricular structure. Circulation Y., Sakamoto, T., Yamamuro, M., Saito, Y., Nakao, K., Yasue, H.,
99, 2255–2260. Ogawa, H., 2002. Expression of aldosterone synthase gene in failing
Seeman, T., Widimsky, J., Hampf, M., Bernhardt, R., 1999. Abolished human heart: quantitative analysis using modified real-time polymerase
nocturnal blood pressure fall in a boy with glucocorticoid-remediable chain reaction. J. Clin. Endocrinol. Metab. 87, 3936–3940.
aldosteronism. J. Hum. Hypertens. 13, 823–828. Young, L.S., Murphy, G., Kelly, S.N., Smith, T.P., Cunningham, S.K.,
Seybert, D.W., 1990. Lipid regulation of bovine cytochrome P450(11) b McKenna, T.J., 2003. Differential production of adrenal steroids by
activity. Arch. Biochem. Biophys. 279, 188–194. purified cells of the human adrenal cortex is relative rather than
Swart, A.C., Brown, N., Kolar, N., Swart, P., 2000. Baboon cytochrome absolute. Eur. J. Endocrinol. 148, 139–145.
P450c11 is encoded by more than one gene. Endocr. Res. 26, 1011– Zuber, M.X., Mason, J.I., Simpson, E.R., Waterman, M.R., 1988. Simul-
1018. taneous transfection of COS-1 cells with mitochondrial and microso-
Takeda, M., Yoneda, T., Demura, M., Mijamori, I., Mabuchi, H., 2000. mal steroid hydroxylases: incorporation of a steroidogenic pathway
Sodium-induced cardic aldosterone synthesis causes cardic hyperten- into nonsteroidogenic cells. Proc. Natl. Acad. Sci. U.S.A. 85, 699–
sion. Endocrinol. 141, 1901–1904. 703.

Das könnte Ihnen auch gefallen