GET THE APP

Congenital Adrenal Hyperplasia, the Origin of Combined Infertilit
Reproductive System & Sexual Disorders: Current Research

Reproductive System & Sexual Disorders: Current Research
Open Access

ISSN: 2161-038X

+44 1300 500008

Case Report - (2014) Volume 3, Issue 4

Congenital Adrenal Hyperplasia, the Origin of Combined Infertility: A Case Report and a Review of Literature

Nardin Aslih1 and Adrian Ellenbogen1,2*
1IVF unit, Hillel Yaffe Medical Center, Hadera, Israel
2Rappaport School of Medicine, Technion – Israel Institue of Technology, Israel
*Corresponding Author: Adrian Ellenbogen, M.D., Clinical Associate Professor, IVF unit, Hillel Yaffe Medical Center, Hadera, Israel, Tel: 972-50-6246722 Email:

Abstract

Congenital Adrenal Hyperplasia (CAH) – a complex and heterogeneous group of conditions is inherited as Autosomal Recessive (AR) disorders. The resultant deficiencies in one of the five enzymes involved in adrenal steroidogenesis lead to defects in the steroidogenic pathways and biosynthesis of cortisol, aldosterone and androgens. Precursor steroids proximal to the blocked step accumulate and can be shunted into other metabolic pathways, particularly that of androgen biosynthesis. CAH due to 21-Hydroxylase deficiency is traditionally separated into two clinical groups: the Classical form (CCAH), which is further separated into salt-wasting (75%) and simple-virilizing (25%) phenotypes, and the Nonclassical form (NC-CAH). They are differentiated by their hormonal profile, predominant clinical features and age of appearance. CAH can affect fertility in females due to inadequate introitus, oligomenorrhea and elevated progesterone levels. Many authors reported an effect on male fertility as well. This editorial describes a case report of combined infertility due to mutations in the CYP21A2 gene and a review of literature on this subject.

<

Keywords: Congenital Adrenal Hyperplasia, CYP21 Deficiency

Case Report

A Couple that presented at our infertility clinic with secondary infertility.

The patient was 33 years old, of Ashkenazi origin, generally healthy. Her gynecologic history included menarche at the age of 15 and oligomenorrhea throughout her life.

Two spontaneous pregnancies with her current partner ended in spontaneous early miscarriages. Duration of infertility at presentation – 6 years. A normal salpingohysterogram ruled out mechanical cause.

Her husband was 43 years old, Jewish of North African origin, generally healthy, a father of 3 children from a previous relationship. Last child conceived with fertility treatments due to male factor. He underwent varicocelle repair few years earlier.

Current sperm analysis revealed severe Oligo-Terato-Asthenospermia (OTA).

Hormonal investigation for causes of oligomenorrhea demonstrated normal FSH, LH, TSH and prolactin levels. 17-OH-progesterone levels were elevated in repeated tests in a range of: 556-2461 ng/dL.

Corticotropin stimulation test was performed; levels of 17-OH-progesterone an hour after stimulation were 1683 ng/dL diagnostic of late onset Congenital Adrenal Hyperplasia (CAH).

Genetic investigation of CYP21 gene mutations revealed that she was homozygous for V281L mutation. Her husband was diagnosed to be a heterozygous carrier for Q318X mutation in the same gene.

Since there was a 50% chance for birth of a child suffering from NC-CAH which is considered a non-lethal disease, there was no indication for Pre-Implantation Genetic Diagnosis (PGD).

The patient conceived in her second IVF cycle treated with an Antagonist protocol. Twelve ova were retrieved, seven were fertilized using micromanipulation, and two good quality fresh embryos were transferred on day 3 resulting in a single embryo pregnancy. Chorionic Villous Sampling (CVS) was done on 12 weeks of gestation confirming she was carrying a healthy female fetus who was a heterozygous carrier of the V281L mutation in the CYP21 gene.

Pregnancy follow up was normal and the patient delivered a healthy baby girl at term.

Introduction

Adrenal gland enlargement was first described by the Napolitan anatomist De Crecchio [1]. The adrenal cortex is formed in the 4th week of gestation, functionally secreting steroids by the 6-7th week of gestation [2]. Biosynthesis of the steroids: cortisol, aldosterone, and androgens from cholesterol occurs under the Adrenocorticotrophic Hormone (ACTH) stimulus with the involvement of five key enzymes : P450scc, 3β-OH dehydrogenase (3β-HSD), 17α -hydroxylase (17α-OH), 21-hydroxylase (21-OH), 11β-hydroxylase (11β-OH) [3,4] (Figure 1).

sexual-disorders-Biosynthesis-steroids

Figure 1: Biosynthesis of steroids.

These steroidogenic enzymes are members of the cytochrome P450 family of oxidases.

Congenital Adrenal Hyperplasia (CAH) – is a complex and heterogeneous group of conditions, inherited as Autosomal Recessive (AR) disorders.

The resultant deficiencies in one of the five enzymes involved in adrenal steroidogenesis lead to defects in the steroidogenic pathways and biosynthesis of cortisol, aldosterone and androgens.

Accumulation of steroid precursors as well as a resulting rise in ACTH levels drives enlargement of the adrenal gland and overproduction of adrenal androgens.

Precursor steroids proximal to the blocked step accumulate and can be shunted into other metabolic pathways, particularly that of androgen biosynthesis. The biochemical and clinical phenotype depends on the specific enzymatic defect and the impairment of specific enzyme activity [5].

All variants are accompanied with glucocorticoid deficiency but each variant of CAH is characterized by a distinct hormonal milieu reflecting the location of the specific block in the path of steroidogenesis.

Defects of the enzymes 21-OH and 11β-OH only affect adrenal steroidogenesis, whereas 17α-OH and 3β-HSD deficiency also impact steroid biosynthesis in the gonads.

The genes of the different CAH forms are well characterized. Confirmation of the diagnosis by genetic analysis is of major clinical significance, as there is a strong genotype–phenotype association for all CAH variants caused by defects in steroidogenenic enzymes [6]. There are five types of CAH (Table 1) [7,8].

Enzyme deficiency Hormone plasma levels Sexual ambiguity
17-OHP DHA Testosterone Aldosterone Renin Males Females
21-OH NC-CAH ↑/N ↑/N N N - -
21-OH C-CAH ↑↑↑ ↑/N ↓/N ↑↑ - +
11β-OH ↑/N ↓↓ ↓↓ - +
3β-HSD ↑↑ Females – N/↑
Males - ↓
↓↓  ↓↓ + ±
17α-OH ↓↓ ↓↓ ↓↓ ↓↓ ↓↓ + -
P450scc (lipoid CAH) 0 0 0 0 ↑↑↑ + -

Table 1: Five types of CAH.
C-CAH: Classical Congenital Adrenal Hyperplasia; NC-CAH: Non-Classical Congenital Adrenal Hyperplasia; 3β-HSD: 3β-OH dehydrogenase; 17α-OH: 17α–hydroxylase; 21-OH: 21-hydroxylase; 11β-OH: 11β-hydroxylase; N:Normal

CAH due to 21-hydroxylase (21-OH) (CYP21A2) deficiency

The most common of the enzyme deficiencies and is responsible of 95% cases of CAH [6]. The resulting disease comprises a complex disease entity with a high degree of heterogenecity depending on the gene mutation and the severity of enzymatic defect (Table 2) [9].

Mutation Severity of enzyme defect (% enzyme activity) Phenotype
Classical mutations
G110Δ8nt Severe (0) SW
I172N Severe (0) SV
Q318X Severe (0) SW
R356W Severe (0) SW,SV
R483P Severe(1-2) SW
Non-classical mutations
P30L Mild (30-60) NC
V281L Mild (20-50) NC
R339H Mild (20-50) NC
P453S Mild (20-50) NC

Table 2: Common mutations in CYP21A2 gene and their phenotypes. SW: Salt Wasting; SV: Simple Virilizing; NC: Non-Classical.

Genotype: The gene encoding 21-OH is located on chromosome 6p21.3, within the HLA histocompatibility complex and it exists in two highly homologous forms: the active gene and the inactive pseudogene [10-13]. More than 100 mutations have been described (Table 3) [14-24].

New born Ambiguous genitalia, Absent testis, Severe shock, Vomiting,
Diarrhea, Hyponarmia, Hyerpkalemia, solated, clithoromegaly, Isolated labial fusion, Penile enlargement,
Hyperpigmentation, Faliure To Thrive (FTT), Hyperisulinemia, Hypoglycemia
Childhood Premature adrenarche
Precocious puberty (central)
Penile enlargement
Small testes
Tall stature and advanced bone age
Adult Oligomenorrhea, Clitoromegaly, Hirsutism, Male-pattern Alopecia, Acne, Seborrhea, Short stature, Deepened voice
Obesity, Insulin resistance-metabolic syndrome Conflicts in gender identity, Male infertility, Testicular adrenal rest tumours, Adrenocortical tumors

Table 3: Clinical spectrum of 21-OH deficiency.

Prevalence: Incidence of the NC-CAH form is almost 1 : 500–1 : 1,000, and of C-CAH is of about 1 in 9800–16,000 live births in most Caucasian populations [10-12]. Incidence varies among ethnic groups and geographical areas. About 65–75% of the CAH patients are compound heterozygous [6]

As reported in some studies CAH is the most common AR disorder in humans. In certain populations such as Ashkenazi Jews incidence is as high as 1:27 [11].

The two 21-OH genes – CYP21A2 encoding the active 21-OH; and the inactive pseudogene CYP21A1P are located approximately 30kb apart. The major mechanism by which the active gene acquires defects is via transfer of segments from the pseudogene to the active gene [12,13]. Disease severity and phenotype are determined by the degree of deficit in 21-OH activity, which is dictated by the least severe CYP21A2 mutation and type of mutation on the allele determining clinical manifestation [5,6].

Phenotype: CAH due to 21-OH deficiency is traditionally separated to two clinical groups: the Classical form (C-CAH), which is further separated into salt-wasting (75%) and simple-virilizing (25%) phenotypes, and the Non-classical form (NC-CAH). They are differentiated by their hormonal profile, predominant clinical features and age of presentation [5,9] (Figure 2) (Table 1).

sexual-disorders-CAH-differentiation

Figure 2: CAH differentiation.

Different authors found a strong association between genotype and phenotype in cases of homozygocity for severe mutations, especially the salt-wasting form. However, the correlation between the genotype and the virilization phenotype assessed by Prader genital stages is less pronounced [6]. In addition, this association is weaker in cases of compound heterozygotes for two different mutations or those carrying mutations of intermediate severity and as a result prediction of phenotype from genotype tends to become more difficult [25-31].

Clinical presentation: C-CAH represents the most common cause of ambiguous genitalia in 46XX infants with the underlying mechanism being in utero exposure to excessive levels of androgens during critical periods of genital development [5]. The severity of virilization of the external genitalia is traditionally classified according to the Prader staging [32,33]. In classical form of CAH the female fetus is exposed to high levels of adrenal androgens at the critical time of sexual differentiation (9-15 weeks) and is born with virilized external genitalia. Internal female genitalia (uterus, fallopian tubes, and ovaries) are normal.

The salt-wasting form presents with severe renal salt loss as a consequence of aldosterone deficiency that can manifest with a life-threatening salt-losing crisis in the neonatal period.

The salt-wasting form presents with severe renal salt loss as a consequence of aldosterone deficiency that can manifest with a life-threatening salt-losing crisis in the neonatal period.

Interestingly, the degree of salt-wasting does not correlate with the degree of virilization [34].

The simple-virilizing form is characterized by virilization of the external genitalia in newborn females and by hypocortisolism and precocious pseudopuberty due to reactive androgen overproduction in both sexes. Early subtle evidence of C-CAH in males which include hyperpigmentation and penile enlargement may go unnoticed [35].

Adequately treated male patients have normal: pubertal development, testicular function, spermatogenesis and fertility. However, small testes and reduced sperm count can occur in patients as a result of inadequately treated disease. Fertility in males with 21-OH deficiency is frequently impaired due to a high incidence of gonadal adrenal rest tumors. These tumors most probably arise from cells having mixed adrenal and Leydig cell properties [15,36].

Steroidogenic cells of both the adrenal and the gonad obviously derive from a common primordium during embryogenesis. The adrenal-like cells express steroidogenic enzymes such as 11β-OH, 17α-OH or 21-OH and respond to ACTH, however, they do not express Leydig cell specific markers. Such a cell population has not been identified yet in human testis [37]. The tumors express adrenal enzymes and ACTH as well as angiontensin receptors. In theory, the atypical local steroid hormone milieu produced by the tumor in the testis results in oligoazoospermia. Long-acting glucocorticoids may reverse fertility, but testis-sparing surgery may be necessary in cases where testicular function cannot be restored hormonally. Semen conservation should be advocated in late adolescence or early adulthood because of the high incidence of such tumors [38].

In addition to impaired adrenocortical function, patients suffering from 21-OHD show a compromised adrenomedullary function [39]. This is due to developmental defects in the formation of the adrenal medulla, leading to depletion of epinephrine stores and decreased production of metanephrine [40]. It seems that normal cortisol secretion by the adrenal cortex is necessary for adrenomedullary organogenesis, because glucocorticoids stimulate the expression of phenylethanolamine-N-methyltransferase, the enzyme that converts norepinephrine to epinephrine [41]. Epinephrine may play a role not only in hypoglycemia observed in children with CAH, but also in leptin concentration (epinephrine inhibits leptin) and probably in insulin resistance that is observed in CAH cases [38,42].

NC-CAH results from mild to moderate enzyme deficiency. It presents postnatally with signs of hyperandrogenism. Females with the NC form are not virilized at birth and do not present with genital ambiguity. Males with NC-CAH were also reported to suffer from: gonadal dysfunction, oligospermia, subfertility, subtle hyperpigmentation, penile enlargement.

Diagnosis: The diagnosis of classical 21-hydroxylase deficiency is made by detecting significantly high levels of 17-OHP, which is the main substrate of the enzyme. Baseline values are >3,500 ng/dL in severely affected infants as compared to those of normal newborns which are <100 ng/dL [43,44].

In practice, the finding of basal follicular-phase morning serum 17-OHP levels >800ng/dL is considered diagnostic for CAH.

The hormonal diagnosis can be further defined by performing ACTH (cosyntropin) stimulation test. Comparing precursor: product ratios can guide differentiating 21-hydroxylase deficiency from other forms of CAH [7]. Nevertheless, in cases of C-CAH ACTH stimulation test in not absolutely necessary for diagnosis.

In most NC patients, stimulated levels of 17-OHP are >1,500 ng/dL. However, the threshold value between NC and heterozygotes is a matter of discussion [43].

Diagnosis must be based on the increased levels of ACTH-stimulated steroid precursor(s) that accumulate above the enzymatic block, with the exception of lipoid adrenal hyperplasia (P450scc deficiency) in which almost no steroids are produced [7].

Neonatal screening: Earlier diagnosis and treatment of CAH can be lifesaving. This is achievable by the simple method of measuring 17-OHP levels on dried blood samples. The screening process, however, is less reliable among low birth weight or preterm infants.

Neonatal screening for CAH can be falsely negative in the event of neonatal dexamethasone treatment. Finally, neonatal screening must be confirmed by genotyping [7].

Prenatal prevention: In pregnancies with offspring at risk of being affected with CCAH, prenatal diagnosis and treatment should be carried out. If the CAH patient’s partner is a carrier of a severe mutation, placing the fetus at risk for CCAH, the patient should be referred to prenatal diagnosis and treatment with dexamethasone until CVS or amniocentesis is performed. The goal is to prevent virilization in 46XX fetuses. Suppressing the active fetal hypothalamic-pituitary-adrenal axis by administrating dexamethasone to the mother prevents androgen overproduction and inappropriate masculinization. Dexamethasone binds minimally to Cortisol Binding Globulin (CBG) in the mother and escapes inactivation by placental 11β hydroxysteroid dehydrogenase enzyme; thus crosses the placenta and may suppress adrenal androgen overproduction. The recommended dose is 20 µg dexamethasone/kg of weight body/day, divided into three doses [7,8,21,35,45-48].

Treatment: The diagnostic evaluation in 21-OHD has become more sophisticated, the standard medical treatment still consists of the same principles which Lawson Wilkins and his colleagues used back in the 1950s [49]. Treatment with glucocorticoid, with or without mineralocorticoid and salt replacement, is directed to prevent adrenal crises and ensuring normal childhood growth by alleviating hyperandrogenism.

Obtaining levels of 17-OHP between 3 and 10 ng/ml usually indicate the efficacy of treatment [13].

Other Less Frequent Forms of CAH

CAH caused by 11β-hydroxylase (CYP11B1) deficiency

About 5–8% of CAH cases are estimated to be caused by 11β-hydroxylase deficiency. This is equivalent to an incidence of 1 in 100,000–200,000 live births. The highest incidence is reported to occur among Jewish population from Morocco (about 1 in 5000–7000 live births) [6].

11OHD results in decreased cortisol secretion and accumulation of the glucocorticoid precursor 11-deoxycortisol and the mineralocorticoid precursor Deoxycorticosterone (DOC). Since DOC can activate the mineralocorticoid receptor, patients can subsequently suffer from significant hypertension, a hallmark feature of this CAH variant [6]. Accumulated precursors are shifted into the androgen synthesis pathway, leading to hyperandrogenism. Classic 11OHD most frequently results in severe virilization of the external genitalia in newborn females, and precocious pseudopuberty in both sexes [50].

CAH caused by 17α-hydroxylase (CYP17A1) deficiency

Steroid 17α-hydroxylase deficiency (17OHD) is a rare form of CAH which accounts for about 1% of all CAH cases. 17OHD results in both glucocorticoid deficiency and sex steroid deficiency. In addition, the mineralocorticoid precursor’s corticosterone and DOC accumulate, exerting significant, albeit weak, glucocorticoid activity. Therefore, 17OHD does not necessarily manifest with adrenal crisis. However, accumulation of corticosterone and DOC also results in mineralocorticoid excess, causing severe hypokalaemic hypertension. Sex steroid deficiency caused by loss of 17,20-lyase activity results in under virilization in male newborns and in primary amenorrhea in 46XX individuals. There is a lack of pubertal development due to hypergonadotropic hypogonadism in both sexes [51].

CAH caused by 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2) deficiency

CAH due to 3β-hydroxysteroid dehydrogenase type - 2 deficiency represents another rare autosomal recessive CAH variant. Data on population-based incidence are lacking. The clinical spectrum shows a wide variety of disease expression but clinical presentation can be largely divided into salt-wasting and non-salt wasting. Genital virilization among female newborns varies variably and as a result diagnosis could be delayed [52].

P450 oxidoreductase deficiency (ORD)-(Antley–Bixler syndrome)

Recently, mutations in the electron donor enzyme P450 oxidoreductase were identified as the cause of CAH. P450 oxidoreductase deficiency (ORD) has a complex phenotype including two unique features not observed in any other CAH variant, skeletal malformations and severe genital ambiguity in both sexes. Apparent paradox of female virilization despite concurrently low circulating androgens observed in this CAH variant suggests the existence of an alternative pathway in human androgen synthesis [53,54]. The biochemical profile suggests partial deficiency of both 17α-hydroxylase and 21-hydroxylase activities. Typical findings include elevated levels of 17OHP, though not to the extent observed in 21OHD. In contrast to 21OHD, sex steroids are low and there is no mineralocorticoid deficiency. Analysis of serum steroids may be confusing and non-diagnostic of the other CAH subtypes [55,56].

Alternative Treatments, New Approaches

Many advances have been made in the management of 21-hydroxylase deficiency during the past 60 years. Despite these advances, the clinical management of patients with CAH is often complicated by abnormal growth and development, iatrogenic Cushing’s syndrome, inadequately treated hyperandrogenism, and infertility [57].

New treatment approaches to classical CAH represent potential solutions to these unresolved issues. These approaches include for example regimens combining a reduced hydrocortisone dose, an anti-androgen, and an aromatase inhibitor [58].

Another approach aims to improve final height in 21-OHD is a combined treatment with GnRH agonists (GnRHa) and Growth Hormone (GH) together with the standard replacement therapy. This approach does not try to avoid high glucocorticoid doses but tries to improve directly poor growth velocity and central precocious puberty directly. Combination therapy with GH and GnRHa appears to improve final height in 21-OHD, but long term randomized, controlled trials are lacking. Obviously, this regimen treats the side effects of glucocorticoid therapy and does not treat the underlying cause of the disease [56].

Other treatment approaches investigated include combination therapy to block androgen action and inhibit testosterone production, and bilateral adrenalectomy in the most severely affected patients [59]. Adrenalectomy will not improve problems caused by gonadal adrenal rests.

Further, even more experimental or theoretical methods of treatment include: corticotrophin-releasing hormone receptor antagonists to avoid adrenal hyperstimulation, carbenoxolone to increase in vivo cortisol concentrations and gene therapy [60].

The applicability and success of these approaches await the results of current research.

References

  1. DeCrecchio L (1865) Sopra un caso di apparenzi virili in una donna. Morgagni 7: 154-188.
  2. Villee DB (1972) The development of steroidogenesis. Am J Med 53: 533-544.
  3. Brown MS, Kovanen PT, Goldstein JL (1979) Receptor-mediated uptake of lipoprotein-cholesterol and its utilization for steroid synthesis in the adrenal cortex. Recent Prog Horm Res 35: 215-257.
  4. Ghayee HK, Auchus RJ (2007) Basic concepts and recent developments in human steroid hormone biosynthesis. Rev Endocr Metab Disord 8: 289-300.
  5. Huynh T, McGown I, Cowley D, Nyunt O, Leong GM, et al. (2009) The clinical and biochemical spectrum of congenital adrenal hyperplasia secondary to 21-hydroxylase deficiency. Clin Biochem Rev 30: 75-86.
  6. Krone N, Arlt W (2009) Genetics of congenital adrenal hyperplasia. Best Pract Res Clin Endocrinol Metab 23: 181-192.
  7. Forest MG (2004) Recent advances in the diagnosis and management of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Hum Reprod Update 10: 469-485.
  8. White PC, Speiser PW (2000) Congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Endocr Rev 21: 245-291.
  9. Nimkarn S, Lin-Su K, New MI (2009) Steroid 21 hydroxylase deficiency congenital adrenal hyperplasia. Endocrinol Metab Clin North Am 38: 699-718.
  10. Yang SY, Levine LS, Zachmann M, New MI, Prader A, et al. (1978) Mapping of the 21-hydroxylase deficiency gene within the HLA linkage group. Transplant Proc 10: 753-755.
  11. Speiser PW, Dupont B, Rubinstein P, Piazza A, Kastelan A, et al. (1985) High frequency of nonclassical steroid 21-hydroxylase deficiency. Am J Hum Genet 37: 650-667.
  12. White PC, Tusie-Luna MT, New MI, Speiser PW (1994) Mutations in steroid 21-hydroxylase (CYP21). Hum Mutat 3: 373-378.
  13. Trakakis E, Basios G, Trompoukis P, Labos G, Grammatikakis I, et al. (2010) An update to 21-hydroxylase deficient congenital adrenal hyperplasia. Gynecol Endocrinol 26: 63-71.
  14. Hughes IA (2007) Congenital adrenal hyperplasia: a lifelong disorder. Horm Res 68 Suppl 5: 84-89.
  15. Cabrera MS, Vogiatzi MG, New MI (2001) Long term outcome in adult males with classic congenital adrenal hyperplasia. J Clin Endocrinol Metab 86: 3070-3078.
  16. Sugino Y, Usui T, Okubo K, Nagahama K, Takahashi T, et al. (2006) Genotyping of congenital adrenal hyperplasia due to 21-hydroxylase deficiency presenting as male infertility: case report and literature review. J Assist Reprod Genet 23: 377-380.
  17. Bonaccorsi A, Adler I, Figueiredo J (1987) Male infertility due to congenital adrenal hyperplasia: testicular biopsy findings, hormonal evaluation, and therapeutic results in three patients. Fertil Steril 47: 664–670.
  18. Claahsen-van der Grinten HL, Sweep FC, Blickman JG, Hermus AR, Otten BJ (2007) Prevalence of testicular adrenal rest tumours in male children with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Eur J Endocrinol 157: 339-344.
  19. Molitor JT, Chertow BS, Fariss BL (1973) Long-term follow-up of a patient with congenital adrenal hyperplasia and failure of testicular development. Fertil Steril 24: 319-323.
  20. Claahsen-van der Grinten HL, Otten BJ, Sweep FC, Hermus AR (2007) Repeated successful induction of fertility after replacing hydrocortisone with dexamethasone in a patient with congenital adrenal hyperplasia and testicular adrenal rest tumors. Fertil Steril 88: 705, e5–8.
  21. New MI (2006) Extensive clinical experience: nonclassical 21-hydroxylase deficiency. J Clin Endocrinol Metab 91: 4205-4214.
  22. Weise M, Mehlinger S, Drinkard B, Rawson E, Charmandari E, et al. (2004) Patients with classic congenital adrenal hyperplasia have decreased epinephrine reserve and defective glucose elevation in response to high intensity exercise. J Clin Endocrinol Metab 59: 591–597.
  23. Merke D, Fields J, Keil M, Vaituzis K, Chrousos G (2003) Children with classic congenital adrenal hyperplasia have decreased amygdala volume: potential prenatal and postnatal hormonal effects. J Clin Endocrinol Metab 88: 1760–1765.
  24. Trakakis E, Loghis C, Kassanos D (2009) Congenital adrenal hyperplasia because of 21-hydroxylase deficiency. A genetic disorder of interest to obstetricians and gynecologists. Obstet Gynecol Surv 64: 177-189.
  25. Dracopoulou M, Maniati M, Dacou C (2001) The spectrum of molecular defects of the CYP 21 gene in the Hellenic population: variable concordance between genotype and phenotype in the different forms of CAH. J Clin Endocrinol Metab 86: 2845–2848.
  26. Speiser PW, White PC (2003) Congenital adrenal hyperplasia. N Engl J Med 349: 776-788.
  27. Wilson RC1, Mercado AB, Cheng KC, New MI (1995) Steroid 21-hydroxylase deficiency: genotype may not predict phenotype. J Clin Endocrinol Metab 80: 2322-2329.
  28. Rumsby G1, Avey CJ, Conway GS, Honour JW (1998) Genotype-phenotype analysis in late onset 21-hydroxylase deficiency in comparison to the classical forms. Clin Endocrinol (Oxf) 48: 707-711.
  29. Speiser PW (2001) Molecular diagnosis of CYP21 mutations in congenital adrenal hyperplasia: implications for genetic counseling. Am J Pharmacogenomics 1: 101-110.
  30. Deneux C1, Tardy V, Dib A, Mornet E, Billaud L, et al. (2001) Phenotype-genotype correlation in 56 women with nonclassical congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Clin Endocrinol Metab 86: 207-213.
  31. Dolzan V1, Sólyom J, Fekete G, Kovács J, Rakosnikova V, et al. (2005) Mutational spectrum of steroid 21-hydroxylase and the genotype-phenotype association in Middle European patients with congenital adrenal hyperplasia. Eur J Endocrinol 153: 99-106.
  32. Prader A, Gurtner HP (1955) The syndrome of male pseudohermaphrodism in congenital adrenocortical hyperplasia without overproduction of androgens (adrenal male pseudohermaphrodism). Helv Paediatr Acta 10: 397–412.
  33. Ogilvy-Stuart AL, Brain CE (2004) Early assessment of ambiguous genitalia. Arch Dis Child 89: 401-407.
  34. Krone N, Braun A, Roscher AA, Knorr D, Schwarz HP (2000) Predicting phenotype in steroid 21-hydroxylase deficiency? Comprehensive genotyping in 155 unrelated, well defined patients from southern Germany. J Clin Endocrinol Metab 85: 1059–1065.
  35. Merke DP, Bornstein SR (2005) Congenital adrenal hyperplasia. Lancet 365: 2125-2136.
  36. Stikkelbroeck ML, Otten BJ, Pasic A, Jager GJ, Sweep CG, et al. (2001) High prevalence of testicular adrenal rest tumors, impaired spermatogenesis, and Leydig cell failure in adolescent and adult males with congenital adrenal hyperplasia. J Clin Endocrinol Metab 86: 5721–5728.
  37. Val P, Jeays-Ward K, Swain A (2006) Identification of a novel population of adrenal-like cells in the mammalian testis. Dev Biol 299: 250-256.
  38. Riepe FG, Sippell WG (2007) Recent advances in diagnosis, treatment, and outcome of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Rev Endocr Metab Disord 8: 349-363.
  39. Merke DP, Chrousos GP, Eisenhofer G, Weise M, Keil MF, et al. (2000) Adrenomedullary dysplasia and hypofunction in patients with classic 21-hydroxylase deficiency. N Engl J Med 343: 1362-1368.
  40. Bornstein SR, Tajima T, Eisenhofer G, Haidan A, Aguilera G (1999) Adrenomedullary function is severely impaired in 21-hydroxylase-deficient mice. FASEB J 13: 1185-1194.
  41. Evinger MJ, Towle AC, Park DH, Lee P, Joh TH (1992) Glucocorticoids stimulate transcription of the rat phenylethanolamine N-methyltransferase (PNMT) gene in vivo and in vitro. Cell Mol Neurobiol 12: 193-215.
  42. Riepe FG, Krone N, Kruger SN, Sweep FC, Lenders JW, et al. (2006) Absence of exercise-induced leptin suppression associated with insufficient epinephrine reserve in patients with classic congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Exp Clin Endocrinol Diabetes 114: 105–110.
  43. New MI, Lorenzen F, Lerner AJ, Kohn B, Oberfield SE, et al. (1983) Genotyping steroid 21-hydroxylase deficiency: hormonal reference data. J Clin Endocrinol Metab 57: 320-326.
  44. Witchel SF, Nayak S, Suda-Hartman M, Lee PA (1997) Newborn screening for 21-hydroxylase deficiency: results of CYP21 molecular genetic analysis. J Pediatr 131: 328-331.
  45. Ogilvie CM, Crouch NS, Rumsby G, Creighton SM, Liao LM, et al. (2006) Congenital adrenal hyperplasia in adults: a review of medical, surgical and psychological issues. Clin Endocrinol (Oxf) 64: 2-11.
  46. Arlt W, Krone N (2007) Adult consequences of congenital adrenal hyperplasia. Horm Res 68 Suppl 5: 158-164.
  47. Nimkarn S, New MI (2007) Prenatal diagnosis and treatment of congenital adrenal hyperplasia. Horm Res 67: 53-60.
  48. Nimkarn S, New MI (2009) Prenatal diagnosis and treatment of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Mol Cell Endocrinol 300: 192-196.
  49. WILKINS L, LEWIS RA, KLEIN R, ROSEMBERG E (1950) The suppression of androgen secretion by cortisone in a case of congenital adrenal hyperplasia. Bull Johns Hopkins Hosp 86: 249-252.
  50. Speiser PW, White PC, Dupont J, Zhu D, Mercado AB, et al. (1994) Prenatal diagnosis of congenital adrenal hyperplasia due to 21-hydroxylase deficiency by allele-specific hybridization and Southern blot. Hum Genet 93: 424-428.
  51. Auchus RJ (2001) The genetics, pathophysiology, and management of human deficiencies of P450c17. Endocrinol Metab Clin North Am 30: 101-119, vii.
  52. Simard J, Ricketts ML, Gingras S, Soucy P, Feltus FA, et al. (2005) Molecular biology of the 3beta-hydroxysteroid dehydrogenase/delta5-delta4 isomerase gene family. Endocr Rev 26: 525-582.
  53. Krone N, Dhir V, Ivison HE, Arlt W (2007) Congenital adrenal hyperplasia and P450 oxidoreductase deficiency. Clin Endocrinol (Oxf) 66: 162-172.
  54. Arlt W, Walker EA, Draper N, Ivison HE, Ride JP, et al. (2004) Congenital adrenal hyperplasia caused by mutant P450 oxidoreductase and human androgen synthesis: analytical study. Lancet 363: 2128-2135.
  55. Shackleton C, Marcos J, Malunowicz EM, Szarras-Czapnik M, Jira P, et al. (2004) Biochemical diagnosis of Antley-Bixler syndrome by steroid analysis. Am J Med Genet A 128A: 223-231.
  56. Adachi M, Asakura Y, Tachibana K, Shackleton C (2004) Abnormal steroidogenesis in three patients with Antley–Bixler syndrome: apparent decreased activity of 17a-hydroxylase, 17,20- lyase and 21-hydroxylase. Pediatrics International 46: 583–589.
  57. White PC, Speiser PW (2002) Long-term consequences of childhood-onset congenital adrenal hyperplasia. Best Pract Res Clin Endocrinol Metab 16: 273-288.
  58. Lin-Su K, Vogiatzi MG, Marshall I, Harbison MD, Macapagal MC (2005) Treatment with growth hormone and luteinizing hormone releasing hormone analog improves final adult height in children with congenital adrenal hyperplasia. J Clin Endocrinol Metab 90: 3318–3325.
  59. Gunther DF, Bukowski TP, Ritzén EM, Wedell A, Van Wyk JJ (1997) Prophylactic adrenalectomy of a three-year-old girl with congenital adrenal hyperplasia: pre- and postoperative studies. J Clin Endocrinol Metab 82: 3324-3327.
  60. Merke DP, Bornstein SR, Avila NA, Chrousos GP (2002) NIH conference. Future directions in the study and management of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Ann Intern Med 136: 320-334.
Citation: Aslih N, Ellenbogen A (2014) Congenital Adrenal Hyperplasia, the Origin of Combined Infertility: A Case Report and a Review of Literature. Reprod Syst Sex Disord 3: 142.

Copyright: © 2013 Aslih N, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top