ArticlePDF Available

Non-classical 11β-hydroxylase deficiency caused by compound heterozygous mutations: A case study and literature review

Authors:

Abstract and Figures

Background: 11β-hydroxylase deficiency (11OHD) is extremely rare, and reports of non-classical 11OHD are even rarer. Non-classical 11OHD usually presents as premature adrenarche, hyperandrogenism, menstrual disorders, and hypertension. Because the symptoms of non-classical 11OHD are mild, delayed diagnosis or misdiagnosis as polycystic ovary syndrome or primary hypertension is common. Case presentation: This paper introduces a case of a young female patient presenting hypertension and menstrual disorders. Laboratory examination revealed increased androgen levels, mild adrenal hyperplasia, mild left ventricular hypertrophy, and mild sclerosis of the lower limb arteries. 11OHD was confirmed by genetic testing, and the patient was found to carry compound heterozygous mutations in CYP11B1 (c.583 T > C and c.1358G > A). The mutation Y195H is located in exon 3 and has not been reported previously. In silico studies indicated that this mutation may cause reduced enzymatic activity. After treatment with hydrocortisone and spironolactone, blood pressure was brought under good control, and menstruation returned to normal. We also conducted a retrospective review of previously reported cases in the literature (over 170 cases since 1991). Conclusions: Early diagnosis of non-classical 11OHD is difficult because its symptoms are mild. The possibility of this disease should be considered in patients with early-onset hypertension, menstrual disorders, and hyperandrogenism to provide early treatment and prevent organ damage due to hypertension and hyperandrogenism. CYP11B1 mutations are known to be race-specific and are concentrated in exons 3 and 8, of which mutations in the former are mostly associated with non-classical 11OHD, whereas mutations in the latter are mostly found in classical 11OHD, characterized by severe loss of enzymatic activity.
Content may be subject to copyright.
C A S E R E P O R T Open Access
Non-classical 11β-hydroxylase deficiency
caused by compound heterozygous
mutations: a case study and literature
review
Dongdong Wang, Jiahui Wang, Tong Tong and Qing Yang
*
Abstract
Background: 11β-hydroxylase deficiency (11OHD) is extremely rare, and reports of non-classical 11OHD are even
rarer. Non-classical 11OHD usually presents as premature adrenarche, hyperandrogenism, menstrual disorders, and
hypertension. Because the symptoms of non-classical 11OHD are mild, delayed diagnosis or misdiagnosis as polycystic
ovary syndrome or primary hypertension is common.
Case presentation: This paper introduces a case of a young female patient presenting hypertension and menstrual
disorders. Laboratory examination revealed increased androgen levels, mild adrenal hyperplasia, mild left ventricular
hypertrophy, and mild sclerosis of the lower limb arteries. 11OHD was confirmed by genetic testing, and the patient
was found to carry compound heterozygous mutations in CYP11B1 (c.583 T > C and c.1358G > A). The mutation Y195H
is located in exon 3 and has not been reported previously. In silico studies indicated that this mutation may cause
reduced enzymatic activity. After treatment with hydrocortisone and spironolactone, blood pressure was brought
under good control, and menstruation returned to normal. We also conducted a retrospective review of previously
reported cases in the literature (over 170 cases since 1991).
Conclusions: Early diagnosis of non-classical 11OHD is difficult because its symptoms are mild. The possibility of this
disease should be considered in patients with early-onset hypertension, menstrual disorders, and hyperandrogenism to
provide early treatment and prevent organ damage due to hypertension and hyperandrogenism. CYP11B1 mutations
are known to be race-specific and are concentrated in exons 3 and 8, of which mutations in the former are mostly
associated with non-classical 11OHD, whereas mutations in the latter are mostly found in classical 11OHD,
characterized by severe loss of enzymatic activity.
Keywords: 11β-hydroxylase deficiency, Genetic testing, Hypertension, Protein function prediction
Background
Congenital adrenal hyperplasia (CAH) is a common genetic
endocrine metabolic disorder, of which 21-hydroxylase
deficiency (21OHD) is the most common type, accounting
for 9099% of all CAH cases. The second most common
type of CAH is 11β-hydroxylase deficiency (11OHD), which
accounts for 0.28% of cases [1]. Steroid 11b-hydroxylase
defects lead to reduced conversion of 11-deoxycortisol
(S) and 11-deoxycorticosterone (DOC) to cortisol and
corticosterone, thereby leading to accumulation of the
two steroid precursors mentioned above. In addition,
an increase in metabolic products towards sex steroids
corresponds to the typical clinical presentation including
low renin hypertension, hypokalemia, hyperandrogenemia,
and genital ambiguity in affected females. Current reports
in the literature relevant to 11βOHD mostly involve
classical 11OHD, and there are relatively few reports on
non-classical 11OHD because of its mild symptoms.
* Correspondence: yangq@sj-hospital.org
Obstetrics and Gynecology Department of Shengjing hospital, China Medical
University, Shenyang 110001, Peoples Republic of China
© The Author(s). 2018 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Wang et al. Journal of Ovarian Research (2018) 11:82
https://doi.org/10.1186/s13048-018-0450-8
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Case presentation
The patient was a 23-year-old female (46, XY karyotype)
diagnosed with hypertension (180/120 mmHg) since age
14 and a BMI of 20.8 kg/m
2
. There was no obvious
masculinization, and her parents stated that there were no
obvious abnormalities in vulva development at birth. Anti-
hypertensive drug therapy (nifedipine sustained-release tab-
lets) had been taken continuously, and blood pressure was
controlled to 130140/8090 mmHg. The patient sought
treatment at our hospital due to menstrual disorders. The
patient is the only child of non-consanguineous healthy
parents from Northeast China. The study was approved
by the ethics committees of China Medical University,
and informed consent was obtained from the patient and
her parents.
Clinical examination and testing
Imaging examinations included an ultrasonic cardio-
gram, a colour Doppler ultrasound of the carotid artery
and lower limb arteries, a pelvic colour Doppler ultra-
sound (SSA660A, Toshiba), and a contrast-enhanced
adrenal computer tomography scan (16-slice computer
tomography machine, GE Lightspeed). Laboratory tests
included measurements of serum potassium, natrium,
testosterone, free testosterone, androstenedione, dehydro-
epiandrosterone sulphate, adrenocorticotropic hormone,
cortisol, 17-hydroxyprogesterone, renin, and aldosterone
using chemiluminescence immunoassays and biochemical
assays.
Genetic analysis
Peripheral blood samples from the patient and her par-
ents were collected for gene analysis. Direct sequencing
was performed on all the exons and the exonintron
boundaries of CYP21A2 (NM_000500) and CYP11B1
(NM_000497.3).
In silico analysis
PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2,Protein
ID for CYP11B1 is NP_000488.3 or P15538) and SIFT/
Provean (http://sift.jcvi.org/) were used to predict whether
an amino acid substitution affects protein function.
The alignment in CYP11 families was performed using
CYP11B1 sequences from different species and other
human steroidogenic P450 cytochromes. PolyPhen-2
and DNAMAN software was used for multiple amino
acid sequence alignment. CYP11B2 (PDB entry: 4DVQ.A),
which shares 93.6% sequence identity with CYP11B1,
was selected as the template for model building of
CYP11B1. The structural representations were generated
using PyMOL 2.0.6.
Results
Clinical characteristics and serum hormone levels
The patients blood pressure at admission was 140/
100 mmHg. The laboratory examination results are shown
in Table 1. Blood potassium was normal, and androgen
levels were increased. Adrenal CT indicated mild hyper-
plasia of both adrenal glands. Echocardiography indicated
mild hypertrophy and slight enlargement of the left
ventricle. Vascular ultrasound indicated mild sclerosis
of the arteries in the lower limbs. Ultrasound of the
uterine adnexa did not reveal any abnormality.
Genetic analysis
No mutations in CYP21A2 were found in the patient.
Exons 3 and 8 of CYP11B1 harboured a compound hetero-
zygous mutation (c.583 T > C and c.1358G > A) leading to
the conversion of tyrosine at amino acid position 195 to
histidine (Y195H) and arginine at amino acid position 453
to glutamine (R453Q) (Fig. 1a). Each parent of the patient
carried one of these heterozygous mutations.
Bioinformatics and in silico analysis of Y195H
Homology alignments indicate that the Tyr195 residue
in CYP11B1 is highly conserved among different species,
but compared to other human CYP family members, it is
only the same in CYP11B2 (Table 2). In silico analysis by
both PolyPhen-2 and SIFT/Provean predicted a pathogenic
effect of the novel mutation Y195H. The amino acid
residue Y195 is localized in the E helix (Fig. 1b).
Table 1 Summary of laboratory data for the affected subject
with steroid 11β-hydroxylase deficiency
Parameter Result Reference
range
Basal
level
3 months after treatment
with hydrocortisone
ACTH (pg/ml) 48.9 16.59 7.263.3
Cortisol (nomol/L) 322 391 171536
Aldosterone (ng/ml) 0.13 0.12 0.070.30
Renin (ng/ml) 0.08 0.44 0.936.56
Serum K
+
(mmol/L) 3.64 4.13 3.505.30
Serum Na
+
(mmol/L) 141.2 139.2 137.0147.0
Testosterone (nmol/L) 6.07 0.76 0.692.77
Androstendione (nmol/L) > 35 6.8 2.0910.82
DHEA-S (umol/L) 5.02 3.31 0.9511.67
Free Testosterone (pmol/L) 42.27 13.24 0.7733.03
Estradiol (pmol/L) 202 73.4587
Progestogen (nmol/L) 4.67 0.643.6
17OHP (nmol/L) 11.6 030
ACTH Adrenocorticotropic Hormone, K+ Potassium, DHEA-S
Dehydroepiandrosterone sulfate, 17OHP 17-hydroxyprogesterone
Wang et al. Journal of Ovarian Research (2018) 11:82 Page 2 of 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Intervention and outcome
After diagnosis of non-classical 11OHD was confirmed, the
patient was given hydrocortisone twice a day (hydrocorti-
sone 10 mg in the morning and 5 mg in the afternoon) and
spironolactone (40 mg each day). Her blood pressure
was brought under good control gradually (120/70 mmHg),
and menstruation returned to normal with decreased
androgen levels (Table 1).
Discussion and conclusions
11βOHD is an autosomal recessive genetic disease. Over
100 mutations of CYP11B1 have been reported in the
literature to date. There are more homozygous than
compound heterozygous mutations (69.1% vs. 29.8%),
and there are few single heterozygous mutations (1.1%),
possibly because mutations in other exons have not yet
been discovered [2,3]. In the past, it was believed that
mutations that cause 11βOHD are mostly concentrated
in exons 2, 6, 7, and 8 [2,4,5]. However, our statistics
revealed that most causative mutations are located in
exons 3 and 8 (40%), and that the distribution in the
other exons is actually more average. Patients carrying
mutations in exon 8 account for the highest proportion
among all patients, and patients carrying the R448H mu-
tation are the most numerous (Fig. 2a)[512]. Because
the highly conserved amino acid sequence near C450 is
located in exon 8, the normal structure of this region is
essential for maintaining normal enzymatic activity [2].
Thus, most point mutations in exon 8 result in severe
reduction of enzymatic activity, thereby resulting in classical
11βOHD. Although there are many mutations in exon 3,
most cause a partial reduction in enzymatic activity and
thus result in non-classical 11βOHD (Fig. 2c)[5,7,1323].
Thus, the probability of a mutation appearing in each exon
is similar, but because the mutations reduce enzymatic
activity to different degrees, different severities of clinical
presentation are observed, and there is a high probability of
misidentifying the disease for mutations in certain exons.
The R448H mutation and 8 other mutations are the most
frequently reported and account for approximately 40% of
all cases (Fig. 2b). CYP11B1 mutation shows significant race
specificity. For example, the R448H mutation mentioned
above is common among Moroccan Jews [6]. Tunisians
often carry the two mutations, G379 V and Q356X [24], of
which Q356X is also common among sub-Saharan Africans
and African-Americans [2,3,2426]. The T318 M muta-
tion is most common among Yemenis [2,27,28], and some
Fig. 1 CYP11B1 sequencing results and 3D molecular schematic representation of the mutation site. aCompound heterozygous mutation
(c.583 T > C and c.1358G > A) that leads to the conversion of tyrosine at amino acid position 195 to histidine (Y195H) and arginine at amino acid
position 453 to glutamine (R453Q). bThree-dimensional model structure of CYP11B1. Green, E helix; red, L helix. The side chains of amino acid
residues Y195 (on the E helix) and R453 (on the L helix) are depicted
Table 2 Homology alignments between CYP family members
Protein ID (UniProtKB/Swiss-Prot) Sequence framing Tyr195
P15538 (HUMAN CYP11B1) TLDVQPSIFH YTIEASNLAL
F7GMV0 (Macaca mulatta CYP11B1) TLDVQPSIFH YTIEASNLAL
F6XJ24 (Equus caballus (Horse) CYP11B1) TLDARPSIFH YTIEASNLAL
P51663 (Ovis aries (Sheep) CYP11B1) TLDIAPSVFR YTIEASTLVL
Q29552 (Sus scrofa (Pig) CYP11B1) TLDIKPSIFR YTIEASNLVL
P15150 (Bos taurus (Bovine) CYP11B1) TLDIAPSVFR YTIEASNLVL
Q3TG86 (Mus musculus (Mouse) CYP11B1) SMDFQSSVFN YTIEASHFVL
P15393 (Rattus norvegicus (Rat) CYP11B1) SINIQSNMFN YTMEASHFVI
P19099 (HUMAN CYP11B2) TLDVQPSIFH YTIEASNLAL
P05108 (HUMAN CYP11A1) SGDISDDLFR FAFESITNVI
P05093 (HUMAN CYP17A1) IDNLSKDSLV DLVPWLKIFP
P08686 (HUMAN CYP21A2) SLLTCSIICY LTFGDKIKDD
P11511 (HUMAN CYP19A1) AESLKTHLDR LEEVTNESGY
Wang et al. Journal of Ovarian Research (2018) 11:82 Page 3 of 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
new mutations such as c.53_54insT, G206 V, W260X,
R448P, and H465L are often found in Saudi Arabs [2931].
The R454C mutation has only been reported among the
Chinese [8,10,32].
The case reported in this paper is a patient carrying
the compound heterozygous mutations Y195H and
R453Q located in exons 3 and 8, respectively, which are
mutation hotspots. Of these mutations, R453Q has been
reported in one individual of European descent and 3
Chinese individuals [8,23,33], and is likely to be more
common among the Chinese [33]. The residue R453 is
located in the L-helix and is adjacent to the Cys-pocket
motif. This domain is highly conserved in the P450 family
of enzymes, and causes 11-hydroxylase activity to decrease
to approximately 1% of the wild-type activity [23]. The
Y195H mutation has not been reported previously. Y195
is located in the E helix. Homology alignments indicate
that this amino acid sequence is relatively conserved, and
predictions using PolyPhen-2 and SIFT/Provean indicate
that this mutation may impair protein function. A mutation
in the neighbouring amino acid T196 has been reported
previously (T196A). This mutation can lead to a 3050%
loss in enzymatic activity and can result in non-classical
11OHD [15]. Three-dimensional structural models show
that Y195 and T196 are located in the middle segment of
the E helix and are close to L463-L464 in the L helix, which
is involved in heme binding [34]. This suggests that Y195H
and T196A are similar and may affect enzymatic activity by
indirectly affecting the structure of the L helix, but to a
small degree; it is thus a mutation that causes non-classical
11OHD.
Because the clinical presentation of non-classical
11OHD is atypical and highly variable, its prevalence
may be underestimated and it may be misdiagnosed.
Some patients may be misdiagnosed as having polycystic
ovary syndrome because of the mildly elevated androgen
levels [13,16], and there are patients that only present
with hypertension with mineralocorticoid features [15].
After the ACTH stimulation test, the degree of increase
in 17-hydroxyprogesterone can be used to differentiate
Fig. 2 Characteristics of previously reported CYP11B1 mutations in the literature. aDistribution of CYP11B1 mutations. bThe 8 most common
types of mutations. cEffect of known mutation sites on 11OH enzymatic activity
Wang et al. Journal of Ovarian Research (2018) 11:82 Page 4 of 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
from non-classical 21OHD, but there is still no consensus
on the diagnostic standards for non-classical 11OHD
[4,17,21]. Baseline and stimulated 11-deoxycortisol
measurements, 11β-hydroxylase activity assays, and urinary
steroid profiling using LC-MS/MS are recommended to
avoid missing the diagnosis of non-classical 11OHD
[16,35]. However, 11-deoxycortisol measurement and
ACTH medications are currently unavailable in most
Chinese hospitals, which hinders the diagnosis of 11OHD,
especially non-classical 11OHD. As the cost of genetic diag-
nosis decreases, genetic testing of CYP11B1 in suspected
patients may make the diagnosis of non-classical 11OHD
convenient and accurate.
Non-classical 11OHD patients may develop hypertension
[14,16], but this is highly variable, and its incidence and
extent are still not clear [35]. The patient in our case devel-
oped severe hypertension at an early age, but because her
diagnosis was never confirmed, antihypertensive therapy
was irregular and she developed left ventricular myocardial
hypertrophy and arteriosclerosis-like changes in the lower
limbs, indicating the importance of early diagnosis and
antihypertensive therapy. The use of mineralocorticoid
receptor antagonists such as spironolactone or eplerenone
for antihypertensive therapy is recommended in such
patients [14]. The patient in this case was given spirono-
lactone therapy, which controlled the blood pressure to
normal.
In summary, the present study reports the case of a
Chinese patient with non-classical 11OHD and presenting
with early-onset hypertension, and carrying compound
heterozygous mutations in CYP11B1, of which one muta-
tion is reported for the first time. We also analysed and
summarised over 170 cases that were previously reported
in the literature and found that exons 3 and 8 were
mutation hotspots. Mutations in exon 3 often result in
non-classical 11OHD, whereas mutations in exon 8 more
often result in complete loss of enzymatic activity and
classical 11OHD. The possibility of non-classical 11OHD
should be considered in hypertensive patients with hyper-
androgenism or elevated mineralocorticoids. Such patients
should be carefully identified and given an early diagnosis
and treatment to avoid the adverse outcomes caused by
hyperandrogenism or long-term hypertension.
Availability of data and materials
The datasets obtained and/or analyzed during the current study are available
from the corresponding author on reasonable request.
Authorscontributions
DDW and JHW performed the molecular genetic studies; DDW and TT
participated in the sequence alignment and drafted the manuscript. DDW
participated in the sequence alignment. QY participated in the design of the
study. All authors read and approved the final manuscript.
Ethics approval and consent to participate
The Hospital Ethics Committee of the Shengjing hospital of China Medical
University approved the study.
Consent for publication
The patient and her family provided written informed consent for
publication of their data.
Competing interests
The authors declare that they have no competing interests.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Received: 12 May 2018 Accepted: 26 August 2018
References
1. Bulsari K, Falhammar H. Clinical perspectives in congenital adrenal hyperplasia
due to 11beta-hydroxylase deficiency. Endocrine. 2017;55(1):1936.
2. Curnow KM, Slutsker L, Vitek J, Cole T, Speiser PW, New MI, et al. Mutations
in the CYP11B1 gene causing congenital adrenal hyperplasia and
hypertension cluster in exons 6, 7, and 8. Proc Natl Acad Sci USA. 1993;
90(10):45526.
3. Merke DP, Tajima T, Chhabra A, Barnes K, Mancilla E, Baron J, et al. Novel
CYP11B1 mutations in congenital adrenal hyperplasia due to steroid 11
beta-hydroxylase deficiency. J Clin Endocrinol Metab. 1998;83(1):2703.
4. Nimkarn S, New MI. Steroid 11beta- hydroxylase deficiency congenital
adrenal hyperplasia. Trends Endocrinol Metab. 2008;19(3):969.
5. Geley S, Kapelari K, Johrer K, Peter M, Glatzl J, Vierhapper H, et al. CYP11B1
mutations causing congenital adrenal hyperplasia due to 11 beta-
hydroxylase deficiency. J Clin Endocrinol Metab. 1996;81(8):2896901.
6. White PC, Dupont J, New MI, Leiberman E, Hochberg Z, Rosler A. A mutation
in CYP11B1 (Arg-448----his) associated with steroid 11 beta-hydroxylase
deficiency in Jews of Moroccan origin. J Clin Invest. 1991;87(5):16647.
7. Grigorescu Sido A, Weber MM, Grigorescu Sido P, Clausmeyer S, Heinrich U,
Schulze E. 21-hydroxylase and 11beta-hydroxylase mutations in Romanian
patients with classic congenital adrenal hyperplasia. J Clin Endocrinol
Metab. 2005;90(10):576973.
8. Zhang M, Liu Y, Sun S, Zhang H, Wang W, Ning G, et al. A prevalent and
three novel mutations in CYP11B1 gene identified in Chinese patients with
11-beta hydroxylase deficiency. J Steroid Biochem Mol Biol. 2013;133:259.
9. Dumic K, Yuen T, Grubic Z, Kusec V, Barisic I, New MI. Two novel CYP11B1
gene mutations in patients from two Croatian families with 11 beta
-hydroxylase deficiency. Int J Endocrinol. 2014;2014:185974.
10. Wang X, Nie M, Lu L, Tong A, Chen S, Lu Z. Identification of seven novel
CYP11B1 gene mutations in Chinese patients with 11beta-hydroxylase
deficiency. Steroids. 2015;100:116.
11. Nguyen HH, Eiden-Plach A, Hannemann F, Malunowicz EM, Hartmann MF,
Wudy SA, et al. Phenotypic, metabolic, and molecular genetic
characterization of six patients with congenital adrenal hyperplasia caused
by novel mutations in the CYP11B1 gene. J Steroid Biochem Mol Biol. 2016;
155(Pt A):12634.
12. Yurekli BS, Kutbay NO, Onay H, Simsir IY, Kocabas GU, Erdogan M, et al. A
novel CYP11B1 mutation in a Turkish patient with 11beta-hydroxylase
deficiency: an association with the severe hypokalemia leading to
rhabdomyolysis. Hormones (Athens, Greece). 2016;15(2):3002.
13. Joehrer K, Geley S, Strasser-Wozak EM, Azziz R, Wollmann HA, Schmitt K, et
al. CYP11B1 mutations causing non-classic adrenal hyperplasia due to 11
beta-hydroxylase deficiency. Hum Mol Genet. 1997;6(11):182934.
14. Mooij CF, Parajes S, Rose IT, Taylor AE, Bayraktaroglu T, Wass JA, et al.
Characterization of the molecular genetic pathology in patients with
11beta-hydroxylase deficiency. Clin Endocrinol. 2015;83(5):62935.
15. Barr M, MacKenzie SM, Wilkinson DM, Holloway CD, Friel EC, Miller S, et al.
Functional effects of genetic variants in the 11beta-hydroxylase (CYP11B1)
gene. Clin Endocrinol. 2006;65(6):81625.
16. Reisch N, Hogler W, Parajes S, Rose IT, Dhir V, Gotzinger J, et al. A diagnosis
not to be missed: nonclassic steroid 11beta-hydroxylase deficiency
presenting with premature adrenarche and hirsutism. J Clin Endocrinol
Metab. 2013;98(10):E16205.
17. Parajes S, Loidi L, Reisch N, Dhir V, Rose IT, Hampel R, et al. Functional
consequences of seven novel mutations in the CYP11B1 gene: four
mutations associated with nonclassic and three mutations causing classic
11{beta}-hydroxylase deficiency. J Clin Endocrinol Metab. 2010;95(2):77988.
Wang et al. Journal of Ovarian Research (2018) 11:82 Page 5 of 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
18. Krone N, Grischuk Y, Muller M, Volk RE, Grotzinger J, Holterhus PM, et al.
Analyzing the functional and structural consequences of two point
mutations (P94L and A368D) in the CYP11B1 gene causing congenital
adrenal hyperplasia resulting from 11-hydroxylase deficiency. J Clin
Endocrinol Metab. 2006;91(7):26828.
19. Menabo S, Polat S, Baldazzi L, Kulle AE, Holterhus PM, Grotzinger J, et al.
Congenital adrenal hyperplasia due to 11-beta-hydroxylase deficiency:
functional consequences of four CYP11B1 mutations. Eur J Hum Genet:
EJHG. 2014;22(5):6106.
20. Krone N, Riepe FG, Gotze D, Korsch E, Rister M, Commentz J, et al.
Congenital adrenal hyperplasia due to 11-hydroxylase deficiency: functional
characterization of two novel point mutations and a three-base pair
deletion in the CYP11B1 gene. J Clin Endocrinol Metab. 2005;90(6):372430.
21. Riedl S, Nguyen HH, Clausmeyer S, Schulze E, Waldhauser F, Bernhardt R. A
homozygous L299P mutation in the CYP11B1 gene leads to complete
virilization in 46,XX individuals with 11-beta-hydroxylase deficiency. Horm
Res. 2008;70(3):1459.
22. Kuribayashi I, Nomoto S, Massa G, Oostdijk W, Wit JM, Wolffenbuttel BH, et
al. Steroid 11-beta-hydroxylase deficiency caused by compound
heterozygosity for a novel mutation, p.G314R, in one CYP11B1 allele, and a
chimeric CYP11B2/CYP11B1 in the other allele. Horm Res. 2005;63(6):28493.
23. Krone N, Grotzinger J, Holterhus PM, Sippell WG, Schwarz HP, Riepe FG.
Congenital adrenal hyperplasia due to 11-hydroxylase deficiency--insights
from two novel CYP11B1 mutations (p.M92X, p.R453Q). Horm Res. 2009;
72(5):2816.
24. Kharrat M, Trabelsi S, Chaabouni M, Maazoul F, Kraoua L, Ben Jemaa L, et al.
Only two mutations detected in 15 Tunisian patients with 11beta-
hydroxylase deficiency: the p.Q356X and the novel p.G379V. Clin Genet.
2010;78(4):398401.
25. Skinner CA, Rumsby G, Honour JW. Single strand conformation
polymorphism (SSCP) analysis for the detection of mutations in the
CYP11B1 gene. J Clin Endocrinol Metab. 1996;81(6):238993.
26. Andrew M, Barr M, Davies E, Wallace AM, Connell JM, Ahmed SF. Congenital
adrenal hyperplasia in a Nigerian child with a novel compound
heterozygote mutation in CYP11B1. Clin Endocrinol. 2007;66(4):6023.
27. Cerame BI, Newfield RS, Pascoe L, Curnow KM, Nimkarn S, Roe TF, et al.
Prenatal diagnosis and treatment of 11beta-hydroxylase deficiency
congenital adrenal hyperplasia resulting in normal female genitalia. J Clin
Endocrinol Metab. 1999;84(9):312934.
28. Motaghedi R, Betensky BP, Slowinska B, Cerame B, Cabrer M, New MI, et al.
Update on the prenatal diagnosis and treatment of congenital adrenal
hyperplasia due to 11beta-hydroxylase deficiency. J Pediatr Endocrinol
Metab : JPEM. 2005;18(2):13342.
29. Bin-Abbas B, Al-Humaida D, Al-Sagheir A, Qasem E, Almohanna M, Alzahrani
AS. Divergent gender identity in three siblings with 46XX karyotype and
severely virilizing congenital adrenal hyperplasia caused by a novel CYP11B1
mutation. Endocr Pract: J Am Coll Endocrinol Am Assoc Clin Endocrinol.
2014;20(10):e1917.
30. Alqahtani MA, Shati AA, Zou M, Alsuheel AM, Alhayani AA, Al-Qahtani SM,
et al. A novel mutation in the CYP11B1 gene causes steroid 11beta-
hydroxylase deficient congenital adrenal hyperplasia with reversible
cardiomyopathy. Int J Endocrinol. 2015;2015:595164.
31. Alzahrani AS, Alswailem M, Murugan AK, Al-Humaida D, Capper CP, Auchus
RJ, et al. A high rate of novel CYP11B1 mutations in Saudi Arabia. J Steroid
Biochem Mol Biol. 2017;174:21724.
32. Wu C, Zhou Q, Wan L, Ni L, Zheng C, Qian Y, et al. Novel homozygous p.
R454C mutation in the CYP11B1 gene leads to 11beta-hydroxylase
deficiency in a Chinese patient. Fertil Steril. 2011;95(3):1122.e36.
33. Gu C, Tan H, Yang J, Lu Y, Ma Y. Congenital adrenal hyperplasia due to 11-
hydroxylase deficiency-compound heterozygous mutations of a prevalent
and two novel CYP11B1 mutations. Gene. 2017;626:8994.
34. Otyepka M, Skopalik J, Anzenbacherova E, Anzenbacher P. What common
structural features and variations of mammalian P450s are known to date?
Biochim Biophys Acta. 2007;1770(3):37689.
35. Escobar-Morreale HF, Carmina E, Dewailly D, Gambineri A, Kelestimur F,
Moghetti P, et al. Epidemiology, diagnosis and management of hirsutism: a
consensus statement by the androgen excess and polycystic ovary
syndrome society. Hum Reprod Update. 2012;18(2):14670.
Wang et al. Journal of Ovarian Research (2018) 11:82 Page 6 of 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Congenital adrenal hyperplasia (CAH) is a genetic endocrine metabolic disease characterized by autosomal recessive defects in one of the adrenal enzymes responsible for glucocorticoid biosynthesis [1,2]. It was first discovered by De Crecchio in 1865 [3]. ...
... CAH should be considered as a spectrum of phenotypes, ranging from asymptomatic to severe [11]. The enzymatic deficiency of 11β-hydroxylase reduces the conversion of 11-deoxycortisol (S) and 11-deoxycorticosterone (DOC) to cortisol and corticosterone, leading to their accumulation and shunting into androgens [1]. Classic 11βOHD presents with features of hyperandrogenism, such as virilization of the external genitalia in female newborns, peripheral precocious puberty, and advanced bone age due to premature epiphyseal closure [1,5,7]. ...
... The enzymatic deficiency of 11β-hydroxylase reduces the conversion of 11-deoxycortisol (S) and 11-deoxycorticosterone (DOC) to cortisol and corticosterone, leading to their accumulation and shunting into androgens [1]. Classic 11βOHD presents with features of hyperandrogenism, such as virilization of the external genitalia in female newborns, peripheral precocious puberty, and advanced bone age due to premature epiphyseal closure [1,5,7]. Moreover, elevation in the levels of mineralocorticoid-like precursors, such as DOC, leads to the development of mild to moderate hyporeninemic hypertension in two-thirds of the cases at the time of diagnosis, sodium retention, hypokalemia, and acidosis [12][13][14][15]; other features include hirsutism, acne, and hyperpigmentation [16]. ...
Article
Full-text available
Congenital adrenal hyperplasia (CAH) is an uncommon condition and 11β-hydroxylase deficiency (11βOHD) accounts for 0.2-8% of cases. In this study, we report a three-year-old girl with a known diagnosis of classical CAH on maintenance treatment with hydrocortisone who presented with abnormal genitalia and persistent hypertension. Genetic testing confirmed the diagnosis of autosomal recessive CAH due to 11βOHD as a result of a novel homozygous pathogenic mutation, c.53dup p.(Gln19Alafs*21), in the CYP11B1 gene. Physicians should consider the possibility of classical 11βOHD in CAH patients presenting with persistent hypertension, even if other laboratory biomarkers are equivocal.
... Treatment of CAH patients requires the participation of a team with an endocrinologist, urologist, gynecologist, psychiatrist, and geneticist (10). Typical manifestations of this disease include low-renin hypertension, hypokalemia, hyperandrogenism, and sexual ambiguity in females (11). Approximately two-thirds of patients with 11βOHD suffer from hypertension that arises in the early years of life, as a result of deoxycorticosterone accumulation in the zona fasciculata and mineralocorticoid effects. ...
... Following the non-classical 11OHD diagnosis, the patient was given hydrocortisone and spironolactone treatment. The patient's blood pressure was well-controlled and the masculinization symptoms reverted to normal with a decrease in androgen levels (11). In Dittmann's report, the cognitive-developmental delay was observed in patients with 11βOHD. ...
Article
Full-text available
Background: 11beta-hydroxylase deficiency (11βOHD) is clinically presented with external genitalia virilization in girls and precocious puberty in boys. Low renin hypertension occurs in both sexes. Early diagnosis and treatment of hypertension can prevent complications. Objectives: To provide a practical management for controlling hypertension in patients with 11-beta-hydroxylase deficiency. Methods: We described a 4.5 years old girl of 46.XX, who presented with ambiguous genitalia at birth and hypertension later in follow-up. The patient had received the appropriate dosage of hydrocortisone and the level of 17-hydroxy progesterone was within the acceptable range but the hypokalemia persisted. Both hypertension and hypokalemia were normalized when spironolactone was added. Results: Blocking mineralocorticoid receptor treat hypertension in 11-beta-hydroxylase deficiency. Conclusions: Intermittent measurement of blood pressure is necessary for patients with 11βOHD. In these patients, spironolactone is effective in treating mineralocorticoid-mediated hypertension and hypokalemia by blocking mineralocorticoid receptors.
... Typically, patients with 11β-OHD exhibit single nucleotide variations (SNVs) and insertions-deletions (indels) in CYP11B1. The primary variant types are missense and loss-of-function variants, both of which can lead to reduced 11β-hydroxylase enzyme activity and disease onset (7,8). ...
Article
In terms of prevalence, 11β-hydroxylase deficiency (11β-OHD), a common form of congenital adrenal hyperplasia, closely follows 21-hydroxylase deficiency. 11β-OHD has been attributed to diminished enzymatic activity owing to CYP11B1 gene variants, mainly encompassing single nucleotide variations and insertions-deletions. The involvement of chimeric CYP11B2/CYP11B1 genes in 11β-OHD has been rarely reported. We conducted a genetic investigation on a male infant with generalized pigmentation and abnormal steroid hormone levels. Whole-exome sequencing revealed a heterozygous variant in CYP11B1 inherited from the mother (NM_000497.4: c.1391_1393dup [p.Leu464dup]). Long-range polymerase chain reaction revealed an additional allele, a chimeric CYP11B2/CYP11B1 gene, inherited from the father. The current case report emphasizes the need to consider the occurrence of gene fusion variants in the diagnosis of neonatal or early infantile 11β-OHD.
... Pathogenic variants in the CYP11B1 gene encoding the enzyme 11βOH are the second most common cause of CAH [2,[10][11][12]. To date, more than 100 pathogenic variants have been identified in the CYP11B1 gene with no more than 13 pathogenic variants being responsible for the development of non-classic 11βOHD [3]. ...
Article
Background Congenital adrenal hyperplasia (CAH) is an autosomal recessive disorder caused by pathogenic variants in seven genes involved in the cortisol and aldosterone biosynthetic pathway. The second most common cause, 11β-hydroxylase deficiency (11βOHD), is attributed to pathogenic variants in the CYP11B1 gene encoding for the enzyme 11β-hydroxylase (11βOH).Case presentationA 13-year-old girl was referred to the pediatric endocrinologist due to a syncopal episode. She is the third child of non-consanguineous parents. She presented with premature adrenarche at the age of 6 years and menarche at the age of 12 years. On physical examination, her height was 154.5 cm and weight 50 kg, while she presented with acne, hirsutism, clitoromegaly, and normal blood pressure. Laboratory investigation revealed increased androgen levels and poor cortisol response to the ACTH stimulation test. From the family history, the mother was diagnosed with CAH at the age of 10 years and was under treatment with methylprednisolone. Previous molecular investigation of the CYP21A2 gene was negative. Due to the increased androstenedione levels in the index patient, the suspicion of 11βOH was raised, and she was investigated for 11-deoxycortisol, 11-deoxycorticosterone, and CYP11B1 gene pathogenic variants. The patient and her mother were found to be compound heterozygous for two novel variants of the CYP11B1 gene.Conclusion We present a case of CAH due to compound heterozygosity of two novel pathogenic variants of the CYP11B1 gene, emphasizing the importance of molecular investigation in order to confirm clinical diagnosis and allow proper genetic counseling of the family.
Article
Full-text available
Purpose 11β-hydroxylase deficiency (11β-OHD) constitutes a rare form of congenital adrenal hyperplasia (CAH), typically accounting for ~5–8% of CAH cases. Non-classical 11β-OHD is reported even more rarely and frequently results in misdiagnosis or underdiagnosis due to its mild clinical symptoms. Methods A clinical, biochemical, radiological, and genetic study was conducted on a 9-year-old girl presenting with mild breast development, axillary hair growth, and advanced bone age. Additionally, a comprehensive review and synthesis of the literature concerning 11β-OHD were conducted. Results The patient presented with breast enlargement, axillary hair development, and accelerated growth over the past year. Laboratory tests revealed levels of cortisol, luteinizing hormone, testosterone, and progesterone that were below normal. A gonadotropin-releasing hormone (GnRH) stimulation test suggested the possibility of central precocious puberty. Radiologic examination revealed a 2-year advance in bone age, while bilateral adrenal ultrasonography showed no abnormalities. Her mother exhibited hirsutism, while her father’s physical examination revealed no abnormalities. Whole-exon genetic testing of the child and her parents indicated a heterozygous mutation of c.905_907delinsTT in exon 5 of the 11β-hydroxylase gene (CYP11B1) in the child and her mother. This mutation resulted in a substitution of aspartic acid with valine at amino acid position 302 of the coding protein. This frameshift resulted in a sequence of 23 amino acids, culminating in a premature stop codon (p.Asp302ValfsTer23). A review of the previous literature revealed that the majority of heterozygous mutations in 11β-OHD were missense mutations, occurring primarily in exons 2, 6, 7, and 8. The most common mutation among 11β-OHD patients was the change of Arg-448 to His (R448H) in CYP11B1. Furthermore, bioinformatics analyses revealed that heterozygous mutation of c.905_907delinsTT had deleterious effects on the function of CYP11B1 and affected the stability of the protein, presumably leading to a partial impairment of enzyme activity. The results of the in vitro functional study demonstrated that the missense mutant (p.Asp302ValfsTer23) exhibited partial enzymatic activity. Conclusions We report a novel heterozygous mutation of CYP11B1 (c.905_907delinsTT), enriching the spectrum of genetic variants of CYP11B1. This finding provides a valuable case reference for early diagnosis of non-classical patients with 11β-OHD.
Article
Congenital forms of endocrine hypertension are rare and potentially life-threatening disorders, primarily caused by genetic defects affecting adrenal steroid synthesis and activation pathways. These conditions exhibit diverse clinical manifestations, which can be distinguished by their unique molecular mechanisms and steroid profiles. Timely diagnosis and customized management approach are crucial to mitigate unfavorable outcomes associated with uncontrolled hypertension and other related conditions. Treatment options for these disorders depend on the distinct underlying pathophysiology, which involves specific pharmacological therapies or surgical adrenalectomy in some instances. This review article summarizes the current state of knowledge on the therapeutic management of congenital forms of endocrine hypertension, focusing on familial hyperaldosteronism, congenital adrenal hyperplasia, apparent mineralocorticoid excess, and Liddle syndrome. We provide an overview of the genetic and molecular pathogenesis underlying each disorder, describe the clinical features, and discuss the various therapeutic approaches available and their risk of adverse effects, aiming to improve outcomes in patients with these rare and complex conditions.
Article
Full-text available
Congenital adrenal hyperplasia (CAH) due to 21α-hydroxylase deficiency (21OHD) or 11β-hydroxylase deficiency (11OHD) are congenital conditions with affected adrenal steroidogenesis. Patients with classic 21OHD and 11OHD have a (nearly) complete enzyme deficiency resulting in impaired cortisol synthesis. Elevated precursor steroids are shunted into the unaffected adrenal androgen synthesis pathway leading to elevated adrenal androgen concentrations in these patients. Classic patients are treated with glucocorticoid substitution to compensate for the low cortisol levels and to decrease elevated adrenal androgens levels via negative feedback on the pituitary gland. On the contrary, non-classic CAH (NCCAH) patients have more residual enzymatic activity and do generally not suffer from clinically relevant glucocorticoid deficiency. However, these patients may develop symptoms due to elevated adrenal androgen levels, which are most often less elevated compared to classic patients. Although glucocorticoid treatment can lower adrenal androgen production, the supraphysiological dosages also may have a negative impact on the cardiovascular system and bone health. Therefore, the benefit of glucocorticoid treatment is questionable. An individualized treatment plan is desirable as patients can present with various symptoms or may be asymptomatic. In this review, we discuss the advantages and disadvantages of different treatment options used in patients with NCCAH due to 21OHD and 11OHD.
Article
Full-text available
Background/Objective Nonclassic Congenital Adrenal Hyperplasia (NCCAH) may be overlooked or mistaken for polycystic ovarian syndrome (PCOS). Unlike Congenital Adrenal Hyperplasia (CAH), the enzymatic activities of 21-hydroxylase or 11-β-hydroxylase in NCCAH are not completely lost. In this case, NCCAH presented in a patient with CYP21A2 and CYP11B1 heterozygous mutations, one of which is a variant of unknown significance (VUS) in CYP11B1. Case Presentation A 30-year-old female presented with a chief complaint of irregular menses and hirsutism. Previous medical history was significant for prolactin of 34.7 ng/mL (reference range 2.0-23.0) and a total testosterone serum testosterone level of 77 ng/dL (reference range 25-125, not sex specific) and a 2x3 mm pituitary lesion. An ACTH stimulation test increased 17-OH progesterone from a baseline 444 ng/dL to 837 ng/dL at 60 minutes (baseline female reference range and stimulated reference ranges are 10-300 ng/dL and <1000 ng/dL, respectively). Gene sequencing revealed a heterozygous pathogenic CYP21A2 variant and a heterozygous previously undescribed VUS in CYP11B1. Discussion Unlike CAH, NCCAH presents more subtly and later in life, and salt wasting and hypertension are not typically seen. \Although mutations in CYP11B1 that cause steroid 11-β-hydroxylase deficiency more commonly lead to the CAH phenotype, case have been reported in CYP11B1 mutations leading to NCCAH depending on the location of the mutations. Conclusion This patient’s case demonstrates physical exam and lab findings suggestive of NCCAH. Our case adds to the database of described mutations in CYP11B1 and suggests that heterozygous mutations in two different genes may present phenotypically as NCCAH.
Article
Full-text available
Congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency is a rare autosomal recessive genetic disorder. It is caused by reduced or absent activity of 11β-hydroxylase (CYP11B1) enzyme and the resultant defects in adrenal steroidogenesis. The most common clinical features of 11 beta-hydroxylase deficiency are ambiguous genitalia, accelerated skeletal maturation and resultant short stature, peripheral precocious puberty and hyporeninemic hypokalemic hypertension. The biochemical diagnosis is based on raised serum 11-deoxycortisol and 11-deoxycorticosterone levels together with increased adrenal androgens. More than 100 mutations in CYP11B1 gene have been reported to date. The level of in-vivo activity of CYP11B1 relates to the degree of severity of 11 beta-hydroxylase deficiency. Clinical management of 11 beta-hydroxylase deficiency can pose a challenge to maintain adequate glucocorticoid dosing to suppress adrenal androgen excess while avoiding glucocorticoid-induced side effects. The long-term outcomes of clinical and surgical management are not well studied. This review article aims to collate the current available data about 11 beta-hydroxylase deficiency and its management.
Article
Full-text available
Congenital adrenal hyperplasia is an autosomal recessive disorder caused by the loss of one of five steroidogenic enzymes affecting cortisol synthesis. In deficiency of 11 β- hydroxylase, 11-Deoxycortisol cannot be converted to cortisol and deoxycorticosterone cannot be converted to corticosterone. We present our 11β-hydroxylase deficiency case with hypokalemia and with a novel mutation. A 20-year-old male patient who’d been on steroid replacement treatment for adrenal insufficiency for 18 years was admitted with spasm, dyspnea and syncope attacks. On laboratory examination his potassium level was 1.4 meq/L and creatinine kinase and myoglobin levels were increased indicating rhabdomyolysis. Potassium infusion was started. 11 β- hydroxylase deficiency related congenital adrenal hyperplasia was diagnosed as the patient had adrenal insufficiency, hypertension and hypokalemia. Spironolactone treatment was started in addition to potassium infusion. Potassium levels were back to normal in the follow-up. Hypokalemia-related rhabdomyolysis regressed. In CYP11B1 whole genome sequencing analysis, p.A199P and p.R448H compound heterozygous mutation was detected. Thus, these two compound heterozygous mutations may be associated with the severity of the disease.
Article
Full-text available
Congenital adrenal hyperplasia (CAH) due to steroid 11β-hydroxylase deficiency is the second most common form of CAH, resulting from a mutation in the CYP11B1 gene. Steroid 11β-hydroxylase deficiency results in excessive mineralcorticoids and androgen production leading to hypertension, precocious puberty with acne, enlarged penis, and hyperpigmentation of scrotum of genetically male infants. In the present study, we reported 3 male cases from a Saudi family who presented with penile enlargement, progressive darkness of skin, hypertension, and cardiomyopathy. The elder patient died due to heart failure and his younger brothers were treated with hydrocortisone and antihypertensive medications. Six months following treatment, cardiomyopathy disappeared with normal blood pressure and improvement in the skin pigmentation. The underlying molecular defect was investigated by PCR-sequencing analysis of all coding exons and intron-exon boundary of the CYP11B1 gene. A novel biallelic mutation c.780 G>A in exon 4 of the CYP11B1 gene was found in the patients. The mutation created a premature stop codon at amino acid 260 (p.W260 (∗) ), resulting in a truncated protein devoid of 11β-hydroxylase activity. Interestingly, a somatic mutation at the same codon (c.779 G>A, p.W260 (∗) ) was reported in a patient with papillary thyroid cancer (COSMIC database). In conclusion, we have identified a novel nonsense mutation in the CYP11B1 gene that causes classic steroid 11β-hydroxylase deficient CAH. Cardiomyopathy and cardiac failure can be reversed by early diagnosis and treatment.
Article
Full-text available
Objective: To describe conflicting gender identities in three karyotypically female siblings with congenital adrenal hyperplasia (CAH) caused by a novel mutation in the CYP11B1 gene, who were assigned as males at birth and followed up to adulthood. Methods: We present 3 siblings (16, 14 and 10 years old) who were born with severe genital virilization and raised as males. Clinical examination showed Prader IV to V external genitalia with a stretched penile length of 7 to 11 cm. Adrenocorticotrophic hormone (ACTH) stimulation test showed a stimulated 11 deoxycortisol (11DOC) level of 12,300-18,700 μg/L (normal 0-5 μg/L). Their karyotypes were 46 XX, and they had normal-sized uterus and ovaries on pelvic ultrasound. DNA was isolated from peripheral leukocytes, and polymerase chain reaction (PCR) and direct sequencing revealed a novel CYP11B1 mutation. This mutation leads to a c.53_54 T insertion (c.53_54insT) with frameshift and truncation at c.115 (codon 39) of CYP11B1. Results: Psychological evaluation of the oldest sibling suggested a female gender identity, and she declared herself as female, and female sex was re-assigned after 1 year of psychosocial adjustment. Psychological assessment for the 2 younger siblings and a fourth 46XY sibling with the same condition revealed male gender identities, and they continued their lives as males without significant difficulties. Conclusion: Divergent gender identity was observed in three severely masculinized 46XX siblings with CAH who carried the same CYP11B1 mutation and had comparable postnatal and probably prenatal androgen exposure and environmental circumstances. These cases suggest that the basis of gender identity is more complex than chromosomal, biochemical, and genetic constitution.
Article
Despite ethnic variation, 11 β-hydroxylase deficiency (11-β OHD) has generally been considered the second most common subtype of congenital adrenal hyperplasia (CAH). We report a high rate of novel mutations in this gene from Saudi Arabia. We studied 16 patients with 11-β OHD from 8 unrelated families. DNA was isolated from peripheral blood. The 9 exons and exon-intron boundaries of CYP11B1 were PCR-amplified and directly sequenced. The novel mutations were functionally characterized using subcloning, in vitro mutagenesis, cell transfection and 11-deoxycortisol: cortisol conversion assays. Six mutations were found in these 8 unrelated families. Three of these mutations are completely novel and two have just been recently described from the same population as novel mutations. They include a single nucleotide insertion mutation in codon 18 (c.53_54insT) leading to frameshift and truncation in 4 siblings, a novel mutation (c.1343 G>C, p.448R>P) in 3 unrelated families, a novel mutation (c.1394 A>T, p.465 H>L) in 2 siblings, a novel mutation (c.617 G>T, p.206 G>V) in 1 patient, and a recently described non-sense novel mutation (c. 780G>A, p.260W>X) in another patient. Out of the 6 mutations described in this report, only one mutation (p.Q356X) was reported previously. In vitro functional testing of the 3 missense and nonsense novel mutations revealed complete loss of the 11 hydroxylase activity. We conclude that CYP11B1 deficiency in Saudi Arabia has a unique genotype with a high rate of novel mutations. The novel p. 448R>P mutation is the most common mutation in this highly inbred population.
Article
11β-hydroxylase deficiency (11β-OHD) occurs in about 5–8% of congenital adrenal hyperplasia (CAH). In this study, we identified three CYP11B1 (encoding Cytochrome P450 11B1) heterozygous mutations: c.1358G > C (p.R453Q), c.1229T > G (p.L410R) and c.1231G > T (p.G411C) in a Chinese CAH patient due to classic 11β-OHD. His parents were healthy and respectively carried the prevalent mutation c.1358G > C (p.R453Q), and the two novel mutations c.1229T > G (p.L410R) and c.1231G > T (p.G411C). In vitro expression studies, immunofluorescence demonstrated that wild type and mutant (L410R and G411C) proteins of CYP11B1 were correctly expressed on the mitochondria, and enzyme activity assay revealed the mutant reduced the 11-hydroxylase activity to 10% (P < 0.001) for the conversion of 11β-deoxycortisol to cortisol. Subsequently, three dimensional homology models for the normal and mutant proteins were built by using the x-ray structure of the human CYP11B2 as a template. Interestingly, in the heme binding site I helix, a change from helix to loop in four amino acide took place in the mutant model. In conclusion, this study expands the spectrum of mutations in CYP11B1 causing to 11β-OHD and provides evidence for prenatal diagnosis and genetic counseling. In addition, our results confirm the two novel CYP11B1 mutations led to impaired 11-hydroxylase activity in vitro.
Article
Congenital adrenal hyperplasia (CAH) is an autosomal recessive inherited disorder of steroidogenesis. Steroid 11β-hydroxylase deficiency (11β-OHD) due to mutations in the CYP11B1gene is the second most common form of CAH. In this study, 6 patients suffering from CAH were diagnosed with 11β-OHD using urinary GC-MS steroid metabolomics analysis. The molecular basis of the disorder was investigated by molecular genetic analysis of the CYP11B1 gene, functional characterization of splicing and missense mutations, and analysis of the missense mutations in a computer model of CYP11B1. All patients presented with abnormal clinical signs of hyperandrogenism. Their urinary steroid metabolomes were characterized by excessive excretion rates of metabolites of 11-deoxycortisol as well as metabolites of 11-deoxycorticosterone, and allowed definite diagnosis. Patient 1 carries compound heterozygous mutations consisting of a novel nonsense mutation p.Q102X (c.304C>T) in exon 2 and the known missense mutation p.T318R (c.953C>G) in exon 5. Two siblings (patient 2 and 3) were compound heterozygous carriers of a known splicing mutation c.1200+1G>A in intron 7 and a known missense mutation p.R448H (c.1343G>A) in exon 8. Minigene experiments demonstrated that the c.1200+1G>A mutation caused abnormal pre-mRNA splicing (intron retention). Two further siblings (patient 4 and 5) were compound heterozygous carriers of a novel missense mutation p.R332G (c.994C>G) in exon 6 and the known missense mutation p.R448H (c.1343G>A) in exon 8. A CYP11B1 activity study in COS-1 cells showed that only 11 % of the enzyme activity remained in the variant p.R332G. Patient 6 carried a so far not described homozygous deletion g.2470_5320del of 2850bp corresponding to a loss of the CYP11B1 exons 3 to 8. The breakpoints of the deletion are embedded into two typical 6 base pair repeats (GCTTCT) upstream and downstream of the gene. Experiments analyzing the influence of mutations on splicing and on enzyme function were applied as complementary procedures to genotyping and provided a rational basis for understanding the clinical phenotype of CAH.
Article
Steroid 11β-hydroxylase (CYP11B1) deficiency (11OHD) is the second most common form of congenital adrenal hyperplasia. Non-classic or mild 11OHD appears to be a rare condition. Our study assessed the residual CYP11B1 function of detected mutations, adding to the spectrum of mild 11OHD and illustrates the variability of the clinical presentation of 11OHD. Five patients presented with mild to moderate 11OHD. Two females presented in adult life with mild hirsutism and in one case with secondary amenorrhoea. Two boys presented with precocious pseudopuberty, gynaecomastia, and elevated blood pressure. One 46,XX female patient was diagnosed with virilisation of the external genitalia two years after birth. Direct DNA sequencing was carried out to perform CYP11B1 mutation analysis. The CYP11B1 mutations were functionally characterized using an in vitro expression system. CYP11B1-inactivating mutations were detected in all patients. Two novel missense mutations (p.P42L and p.A297V) and the previously characterized p.R143W mutation had residual CYP11B1 activities between 10 to 27%. A novel p.L382R and the previously uncharacterized p.G444D mutation both caused complete loss of CYP11B1 enzymatic activity. Mutations causing partial impairment of 11β-hydroxylase activity (residual activity of 10% or above) are associated with a less severe clinical presentation of 11OHD, which can be classified as a non-classic form. Our data demonstrates that patients with non-classic 11OHD can present with androgen excess, precocious pseudopuberty and increased blood pressure. Timely diagnosis of non-classic 11OHD and consequently initiation of personalized treatment is essential to prevent co-morbidities caused by androgen excess and hypertension. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Article
Steroid 11β-hydroxylase deficiency (11β-OHD), one of common cause of congenital adrenal hyperplasia (CAH), is an autosomal recessive disorder characterized by virilization, precocious pseudo-puberty, and hypertension. It is caused by CYP11B1 gene mutation. We performed molecular genetic analysis of the CYP11B1 gene in six patients with preliminary clinical diagnosis of 11β-OHD and four patients identified as potential 11β-OHD from a CAH cohort in which CYP21A2 gene mutations consecutively screened. Seven novel CYP11B1 mutations, including p.R454H, p.Q472P, p.Q155X, p.K173X, IVS2-1G>A, R454A fs 573X, and g.2704_g.3154del, and six previously described mutations (p.P94L, p.G267S, p.G379V, p.R448H, p.R454C and p.R141X) were identified. These mutations mainly clustered in exons 3 and 8. Eight of twenty alleles carried mutations occurring at the Arg454 position, which is a mutational hot spot for Han Chinese. The pathogenic nature of novel p.R454H mutation was predicted by protein sequence alignment and in silico analysis. All the identified mutations were responsible for the clinical features observed in these ten unrelated Chinese patients. This study expands the CYP11B1 mutation spectrum and provides evidence for prenatal diagnosis and genetic counseling. Genetic analysis is an alternative approach to help clinicians confirm uncertain 11β-OHD diagnosis, facilitating reasonable steroid replacement. Copyright © 2015. Published by Elsevier Inc.