Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Horm Res Paediatr ; 77(2): 100-7, 2012.
Article in English | MEDLINE | ID: mdl-22456342

ABSTRACT

BACKGROUND/AIM: X-linked adrenal hypoplasia congenita (AHC) is a rare disorder characterized by primary adrenal insufficiency and hypogonadic hypogonadism. It is caused by deletions or point mutations of the NR0B1 gene, on Xp21. AHC can be associated with glycerol kinase deficiency, Duchenne muscular dystrophy and mental retardation (MR), as part of a contiguous gene deletion syndrome. A synthetic probe set for multiplex ligation-dependent probe amplification analysis was developed to confirm and characterize NR0B1 deletions in patients with AHC and to correlate their genotypes with their divergent phenotypes. RESULTS: In 2 patients, isolated AHC was confirmed, while a patient at risk for metabolic crisis was revealed as the deletion extends to the GK gene. A deletion extending to IL1RAPL1 was confirmed in both patients showing MR. Thus, a good genotype-phenotype correlation was confirmed. CONCLUSIONS: Multiplex ligation-dependent probe amplification analysis is a valuable tool to detect NR0B1 and contiguous gene deletions in patients with AHC. It is especially helpful for IL1RAPL1 deletion detection as no clinical markers for MR are available. Furthermore, multiplex ligation-dependent probe amplification has the advantage to identify female carriers that, depending on the deletion extension, have a high risk of giving birth to children with MR, AHC, glycerol kinase deficiency and Duchenne muscular dystrophy.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/physiopathology , DAX-1 Orphan Nuclear Receptor/genetics , Gene Deletion , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/physiopathology , Genetic Loci , Adolescent , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Insufficiency , Adult , Child , DAX-1 Orphan Nuclear Receptor/metabolism , Female , Genetic Association Studies , Genetic Carrier Screening/methods , Genetic Diseases, X-Linked/metabolism , Humans , Hypoadrenocorticism, Familial , Infant , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Male , Mental Retardation, X-Linked/genetics , Mothers , Multiplex Polymerase Chain Reaction , Nucleic Acid Amplification Techniques , Oligonucleotide Array Sequence Analysis , Sweden
2.
Horm Res Paediatr ; 74(5): 358-64, 2010.
Article in English | MEDLINE | ID: mdl-20606393

ABSTRACT

BACKGROUND: Relative adrenocortical insufficiency is often seen in sick premature newborns. As the human fetal adrenal cortex does not express the 3ß-hydroxysteroid dehydrogenase (3ß-HSD) enzyme before about 23 weeks of gestation, we hypothesized that this enzymatic step may be rate limiting in cortisol synthesis in premature infants of less than 28 weeks postmenstrual age at birth. METHODS: We measured cord, first day (D0) and median fourth day (D4) serum 17-OH-pregnenolone (17-OHPreg), 17-OH-progesterone (17-OHProg), 11-deoxycortisol, cortisol (F) and dehydroepiandrosterone sulphate concentrations and calculated the substrate/product ratios in 67 infants with gestational age 23.6-33.1 weeks. RESULTS: The mean 17-OHPreg/17-OHProg ratio as a marker of 3ß-HSD activity did not differ between the gestational age groups (gestational age <28 vs. ≥28 weeks: 0.40 vs. 0.48, p = 0.52 for cord, 3.1 vs. 2.4, p = 0.25 for D0, and 1.6 vs. 1.9, p = 0.62 for D4). In addition, the 17-OHPreg/17-OHProg ratio did not differ between the infants in the lowest F tertile compared to those in the highest F tertile group, and the serum 17-OHPreg and 17-OHProg concentrations were parallel with the respective F concentrations. CONCLUSION: We did not find evidence of significant immaturity in adrenal 3ß-HSD activity in preterm infants between 24 and 28 weeks of gestation.


Subject(s)
Adrenal Cortex Hormones/blood , Adrenal Cortex/growth & development , Adrenal Cortex/physiology , 17-alpha-Hydroxypregnenolone/blood , 17-alpha-Hydroxyprogesterone/blood , 3-Hydroxysteroid Dehydrogenases/metabolism , Adrenal Cortex Function Tests , Adult , Apgar Score , Birth Weight , Chorioamnionitis/pathology , Cohort Studies , Cortodoxone/blood , Dehydroepiandrosterone Sulfate/blood , Female , Fetal Blood/chemistry , Gestational Age , Humans , Hydrocortisone/biosynthesis , Infant, Newborn , Infant, Premature , Infant, Very Low Birth Weight , Male , Pre-Eclampsia/pathology , Pregnancy , Prospective Studies
3.
Horm Res ; 72(5): 281-6, 2009.
Article in English | MEDLINE | ID: mdl-19844114

ABSTRACT

BACKGROUND: Steroid 11-hydroxylase (CYP11B1) deficiency (11OHD) is the second most common form of congenital adrenal hyperplasia (CAH). Herein, we describe two novel CYP11B1 mutations (g659_660dupTG, p.M92X; g.4817G>A, p.R453Q) found in a patient diagnosed with classic 11OHD, after presenting with borderline elevated 17-hydroxyprogesterone concentrations in CAH newborn screening. METHODS: A novel CYP11B1 variant (p.R453Q) identified in a patient with classic 11OHD was characterized employing a COS7 cell assay and a computational three-dimensional CYP11B1 model. RESULTS: The in vitro expression analysis revealed an almost complete loss of function of p.R453Q. This finding was consistent with the clinical presentation of classic 11OHD as the patient was compound heterozygous for p.R453Q and a nonsense mutation (p.M92X) on the other allele. Inserting the p.R453Q mutation into our CYP11B1 model provided two potential explanations for the almost complete loss of enzyme activity. Firstly, the heme coordination is most likely disturbed. A second possibility could be an altered interaction with the redox partner adrenodoxin. CONCLUSION: Results indicate that both novel mutations are disease-causing mutations. Proving the pathogenic effect of a missense sequence variation is of particular importance for clinical genetic counseling as this provides essential information on the prediction of recurrence risk and disease severity.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Mutation, Missense , Steroid 11-beta-Hydroxylase/genetics , Animals , Arginine/genetics , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Mutational Analysis , Glutamic Acid/genetics , Humans , Infant , Male , Methionine/genetics , Models, Molecular , Mutation, Missense/physiology , Pedigree , Polymorphism, Single Nucleotide , Transfection
4.
J Clin Endocrinol Metab ; 94(8): 3058-64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19454579

ABSTRACT

CONTEXT: Steroid 17alpha-hydroxylase (CYP17A1, alias P450c17) deficiency (17OHD) is a rare form of congenital adrenal hyperplasia. The CYP17A1 enzyme catalyzes two distinct reactions, 17alpha-hydroxylase and 17,20-lyase activities. OBJECTIVE: The aim of the study was to analyze the structural and functional consequences of three novel (A174E, V178D, and L465P) and one previously reported (R440C) CYP17A1 mutation found in three patients clinically and biochemically presenting with 17OHD. PATIENTS AND METHODS: Two patients suffering from 46,XY disordered sex development presented at ages 5.5 and 8.8 yr, respectively, with tall stature and hypertension. Mutation analysis revealed compound heterozygous CYP17A1 mutations (A174E/K388X; V178D/R440C). The third patient (46,XX) presented with primary amenorrhea and hypertension at age 15 yr. She was homozygous for the novel L465P mutation. Functional studies employing a yeast microsomal expression system compared wild-type and mutant CYP17A1 both with regard to 17alpha-hydroxylase and 17,20-lyase activity. Mutants were examined in a computational three-dimensional model of the CYP17A1 protein. RESULTS: The activity assays showed that all three mutants retain only 0-7% of both 17alpha-hydroxylase and 17,20-lyase activity relative to CYP17A1 wild-type activity, corresponding to the in vivo situation. Enzyme kinetic studies proved the impairment of both reactions, respectively. Computer-based three-dimensional model analysis of CYP17A1 using CYP2B4 as template showed that three of the mutations had no direct effect on the active center, whereas one affects the heme coordination. CONCLUSION: The functional studies revealed that the described missense mutations result in severe 17OHD. Our data are important to predict the phenotypic expressions and provide important information for patient management and genetic counseling.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Mutation , Steroid 17-alpha-Hydroxylase/genetics , Child , Child, Preschool , Computer Simulation , Female , Humans , Models, Molecular , Steroid 17-alpha-Hydroxylase/chemistry , Steroid 17-alpha-Hydroxylase/physiology
5.
Hum Mutat ; 30(2): E443-50, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19058224

ABSTRACT

Congenital adrenal hyperplasia (CAH) due to steroid 21-hydroxylase (CYP21A2) deficiency is the commonest inborn error in steroid hormone biosynthesis. Functional in vitro assessment of mutant activity generally correlates well with clinical phenotype and therefore has contributed greatly to phenotype prediction in this CAH variant. Three CYP21A2 sequence variants (g.1641C>T, p.A265V; g.1752G>C, p.W302S; and g.2012A>G, p.D322G) identified in patients with non-classic and simple virilizing CAH were characterized using a yeast co-expression system and a computational three-dimensional CYP21A2 model. Computational analysis of the mutants in the three-dimensional structural model predicted no relevant effect of p.A265V, while p.W302S and p.D322G were predicted to impact significantly on enzyme function. Consistent with these findings, in vitro mutant analysis revealed enzyme activity similar to wild-type for p.A265V, whereas p.W302S and p.D322G exerted activities compatible with simple virilizing and non-classical CAH, respectively. The results indicate that p.A265V is an allelic variant rather than a disease-causing amino acid change, whilst p.W302S and p.D322G could be confirmed as functionally relevant mutations. These findings emphasize the value of in vitro functional analysis of sequence variations in predicting genotype-phenotype correlations and disease severity.


Subject(s)
Mutant Proteins/metabolism , Mutation/genetics , Saccharomyces cerevisiae/metabolism , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/metabolism , Adolescent , Child , Female , Humans , Kinetics , Models, Molecular , Protein Biosynthesis , Protein Structure, Secondary , Steroid 21-Hydroxylase/biosynthesis , Transcription, Genetic
6.
J Clin Endocrinol Metab ; 93(7): 2891-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18445671

ABSTRACT

BACKGROUND: Congenital adrenal hyperplasia is caused by insufficient adrenal steroid biosynthesis due to impaired steroidogenic enzymes. The majority of patients suffer from deficiency of 21-hydroxylase (CYP21) coded by the CYP21A2 gene. OBJECTIVE: Our objective was to study the functional and structural consequences of the novel CYP21A2 missense mutation c.364A > C (K121Q) detected in a female patient with nonclassical 21-hydroxylase deficiency. The patient was compound heterozygous for the novel K121Q mutation and the mild P453S mutation. RESULTS: In vitro expression analysis of the mutant K121Q enzyme in transiently transfected COS-7 cells revealed reduced CYP21 activity of 14.0 +/- 5% for the conversion of 17-hydroxyprogesterone and 19.5 +/- 4% for the conversion of progesterone. K121 is located on helix C in the CYP21 protein, which is part of the heme coordinating system. In addition, helix C is involved in the interaction with the electron-providing enzyme P450 oxidoreductase. Protein modeling revealed that the substitution of glutamine for the basic amino acid lysine introduces an electrostatic change on the surface of CYP21 and may additionally change heme coordination. We hypothesize that the electron flux between P450 oxidoreductase and CYP21 is impaired and, moreover, that substrate affinity is altered due to heme dislocation with K121Q. CONCLUSION: Both the interaction of P450 oxidoreductase and CYP21 as well as heme coordination are likely to be disturbed due to the K121Q mutation. Our data exemplify how the combination of in vitro expression and structural protein analysis provide novel insights into molecular mechanisms of reduced CYP21 activity, eventually explaining the patient's phenotype.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , NADPH-Ferrihemoprotein Reductase/chemistry , Point Mutation , Steroid 21-Hydroxylase/genetics , Child , Female , Humans , Models, Molecular , Steroid 21-Hydroxylase/chemistry , Steroid 21-Hydroxylase/metabolism
7.
Rev Endocr Metab Disord ; 8(4): 349-63, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17885806

ABSTRACT

Congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency (21-OHD) is an autosomal-recessive disease causing cortisol deficiency, aldosterone deficiency and hyperandrogenism. Diagnosis of 21-OHD is confirmed by steroid analysis in newborn screening or later on. Standard medical treatment consists of oral glucocorticoid and mineralocorticoid administration in order to suppress adrenal androgens and to compensate for adrenal steroid deficiencies. However, available treatment is far from ideal, and not much is known about the long-term outcome in CAH as trials in patients in adulthood or old age are rare. Here we briefly describe the pathophysiology, clinical picture, genetics and epidemiology of 21-OHD. This is followed by a comprehensive review of the recent advances in diagnosis, treatment and outcome. Novel insights have been gained in the fields of newborn screening, specific steroid measurement utilizing mass spectrometry, genetics, glucocorticoid stress dosing, additive medical therapy, prenatal treatment, side-effects of medical treatment, adrenomedullary involvement, metabolic morbidity, fertility and gender identity. However, many issues are still unresolved, and novel questions, which will have to be answered in the future, arise with every new finding.


Subject(s)
Adrenal Hyperplasia, Congenital/diagnosis , Adrenal Hyperplasia, Congenital/drug therapy , Steroid 21-Hydroxylase/metabolism , Adrenal Hyperplasia, Congenital/enzymology , Humans , Hydrocortisone/therapeutic use , Infant, Newborn , Neonatal Screening , Steroid 21-Hydroxylase/genetics , Treatment Outcome
8.
Eur J Endocrinol ; 156(2): 249-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17287415

ABSTRACT

OBJECTIVE: The renal form of pseudohypoaldosteronism type 1 (PHA1) is a rare disease caused by mutations in the human mineralocorticoid receptor gene (NR3C2). DESIGN: Aim of the study was to analyze the NR3C2 gene in three Italian patients with clinical signs of renal PHA1 and to evaluate the distribution of the -2G > C, c.538A > G, and c.722C > T single nucleotide polymorphism (SNP) pattern in the PHA1 patients and in 90 controls of the same ethnic origin. METHODS: Analysis of the NR3C2 gene sequence and of the polymorphic SNP markers. Functional characterization of the detected novel NR3C2 mutations utilizing aldosterone-binding assays and reporter gene transactivation assays. RESULTS: One novel nonsense (Y134X) and one novel frameshift (2125delA) mutation were detected. They exhibited no aldosterone binding and no transactivation abilities. No mutation was detected in the third patient. Haploinsufficiency of NR3C2 was ruled out by microsatellite analysis in this patient. The c.722T SNP was detected in 97% of alleles in the Italian population which is significantly different from the general German or US population. CONCLUSIONS: Molecular analysis of the NR3C2 gene in PHA1 patients is warranted to detect novel mutations in order to clarify the underlying genetic cause, which may extend the insight into relevant functional regions of the hMR protein. The effect the different distribution of the c.722T SNP is not clear to date. Further studies are necessary to provide evidence as to a possible advantage of a less sensitive hMR in southern countries.


Subject(s)
Pseudohypoaldosteronism/genetics , Receptors, Mineralocorticoid/genetics , Adult , Aldosterone/blood , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Female , Frameshift Mutation , Gene Frequency , Humans , Hydrocortisone/blood , Italy , Male , Pedigree , Polymorphism, Single Nucleotide , Protein Biosynthesis
9.
J Clin Endocrinol Metab ; 91(12): 4976-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16984992

ABSTRACT

CONTEXT: Congenital adrenal hyperplasia is a group of autosomal recessive inherited disorders of steroidogenesis. The most frequent cause is the deficiency of steroid 21-hydroxylase (CYP21) due to mutations in the CYP21A2 gene. OBJECTIVE: We analyzed the functional and structural consequences of the four CYP21A2 missense mutations (C169R, G178R, W302R, and R426C) to prove their clinical relevance and study their impact on CYP21 function. RESULTS: Analyzing the mutations in vitro revealed an almost absent or negligible CYP21 activity for the conversion of 17-hydroxyprogesterone to 11-deoxycortisol and progesterone to deoxycorticosterone. Protein translation and intracellular localization were not affected by the mutants, as could be demonstrated by Western blotting and immunofluorescence studies. Analysis of these mutants in a three-dimensional model structure of the CYP21 protein explained the observed in vitro effects because all the mutations severely interfere either directly or indirectly with important structures of the 21-hydroxylase protein. CONCLUSION: The in vitro expression analysis of residual enzyme function is a complementary method to genotyping and an important tool for improving the understanding of the clinical phenotype of 21-hydroxylase deficiency. This forms the foundation for accurate clinical and genetic counseling and for prenatal diagnosis and treatment. Moreover, this report demonstrates that the combination of in vitro enzyme analysis and molecular modeling can yield novel insights into CYP450 structure-functional relationships.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Mutation, Missense , Steroid 21-Hydroxylase/genetics , Adolescent , Amino Acid Sequence , Animals , COS Cells , Child , Chlorocebus aethiops , DNA Mutational Analysis , Disorders of Sex Development/genetics , Female , Humans , Infant , Male , Models, Molecular , Molecular Sequence Data , Protein Conformation , Puberty, Precocious/genetics , Russia , Sequence Homology, Amino Acid , Structure-Activity Relationship
10.
J Clin Endocrinol Metab ; 91(11): 4552-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16954160

ABSTRACT

CONTEXT: Pseudohypoaldosteronism type 1 (PHA1) is a rare salt-wasting syndrome. Mutations in the NR3C2 gene coding for the mineralocorticoid receptor (MR) cause autosomal dominant PHA1. OBJECTIVE: Our objective was to reveal the cause of renal salt loss in six PHA1 patients and analyze the mutants' functional impact on MR function. DESIGN: Our study included the following: clinical and hormonal characterization of the patients' phenotype, analysis of the NR3C2 gene, determination of receptor affinities to aldosterone and the transcriptional activation abilities of the MR mutants, investigation of subcellular translocation using fluorescence-labeled MR, and studying changes in mutant receptor conformation with proteolysis experiments and three-dimensional modeling. RESULTS: Six heterozygous NR3C2 mutations were detected. One frameshift mutation (c.1131dupT) has been reported previously. The second frameshift mutation (c.2871dupC), which has only recently been reported by our group, showed no aldosterone binding and no transactivation because of a major change in receptor conformation. Two novel nonsense mutations generate a truncated receptor protein. Two missense mutations differently affect MR function. S818L was reported recently without complete in vitro data. S818L does not bind aldosterone or activate transcription or translocate into the nucleus. A major displacement of several residues involved in aldosterone binding was PHA1 causing. The novel E972G mutation showed a significantly lower ligand-binding affinity and only 9% of wild-type transcriptional activity caused by major changes in receptor conformation. CONCLUSIONS: Our data on six mutations extend the spectrum of PHA1-causing NR3C2 gene mutations. Studying naturally occurring mutants helps to clarify their pathogenicity and to identify crucial residues for MR structure and function.


Subject(s)
Mutation , Pseudohypoaldosteronism/etiology , Pseudohypoaldosteronism/genetics , Receptors, Mineralocorticoid/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Expression , Humans , Imaging, Three-Dimensional , Male , Mice , Models, Molecular , Molecular Sequence Data , Mutant Proteins/analysis , Mutant Proteins/metabolism , Pedigree , Protein Structure, Secondary , Protein Transport , Pseudohypoaldosteronism/metabolism , Rabbits , Receptors, Mineralocorticoid/chemistry , Receptors, Mineralocorticoid/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Transcriptional Activation , Transfection
11.
J Dtsch Dermatol Ges ; 4(8): 654-7, 2006 Aug.
Article in German | MEDLINE | ID: mdl-16895568

ABSTRACT

Infantile acne is a rare condition which usually begins after the third month of life and appears mainly on the cheeks. Spontaneous healing typically occurs within 2 years. A 2-year-old boy developed acne infantum during the first days after birth; it failed to respond to topical treatment. Detailed physical examination and endocrinologic evaluation confirmed the presumed diagnosis of congenital adrenal hyperplasia (CAH), and showed it was caused by 11-beta-hydroxylase deficiency. CAH comprises a group of autosomal-recessively inherited disorders. These hereditary enzyme defects in steroid biosynthesis cause glucocorticoid deficiency and an overproduction of biosynthetic precursor steroids. Adrenal androgen biosynthesis is not impaired but shows a massive reactive overproduction due to the increased ACTH secretion within the up-regulated hypothalamo-pituitary-adrenal system. The characteristic features of CAH in male infants are demonstrated on the basis of this case report.


Subject(s)
Acne Vulgaris/diagnosis , Acne Vulgaris/etiology , Adrenal Hyperplasia, Congenital/complications , Adrenal Hyperplasia, Congenital/diagnosis , Steroid 11-beta-Hydroxylase/metabolism , Acne Vulgaris/genetics , Adrenal Hyperplasia, Congenital/genetics , Child, Preschool , Humans , Male
12.
J Clin Endocrinol Metab ; 91(9): 3671-5, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16757525

ABSTRACT

BACKGROUND: The renal form of pseudohypoaldosteronism type 1 (PHA1) is a rare disease characterized by congenital mineralocorticoid resistance of the kidney. Twenty-two different loss-of-function mutations in the mineralocorticoid receptor gene have been described in families with PHA1. These mutations were not recurrent and resulted in a large phenotypic variability. OBJECTIVE: The objective of this study is to analyze the recurrence of an inactivating mutation in the mineralocorticoid receptor gene in unrelated families with autosomal dominant PHA1. PATIENTS: Seventeen members from three unrelated families with autosomal dominant PHA1 were studied, including 11 affected patients with variable clinical manifestations. Fifty healthy subjects were used as controls. METHODS: Genomic DNA was extracted, and the entire coding region of the mineralocorticoid receptor gene was submitted to automatic sequencing. Four dinucleotide microsatellite markers spanning a region of 3.2 cM in the human mineralocorticoid receptor gene locus, and two intragenic polymorphisms were used for haplotype analysis. RESULTS: A heterozygous point mutation at codon 947 (c.2839C>T) changing arginine to stop codon (R947X) was found in the three families. Different haplotypes segregated with the R947X mutation in each family, demonstrating the absence of a founder effect for this mutation. CONCLUSION: Codon 947 of the mineralocorticoid receptor is the first mutational hot spot for autosomal dominant PHA1.


Subject(s)
Point Mutation , Pseudohypoaldosteronism/genetics , Receptors, Mineralocorticoid/genetics , 17-alpha-Hydroxyprogesterone/blood , Aldosterone/blood , DNA/chemistry , DNA/genetics , Female , Humans , Hydrocortisone/blood , Infant , Infant, Newborn , Male , Microsatellite Repeats , Pedigree , Polymerase Chain Reaction , Polymorphism, Genetic , Pseudohypoaldosteronism/blood , Pseudohypoaldosteronism/pathology , Renin/blood
13.
Mol Cell Endocrinol ; 254-255: 217-20, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16757104

ABSTRACT

Depot gonadotropin releasing hormone (GnRH) agonist (GnRHa) therapy is the treatment of choice for patients with central precocious puberty (CPP). It is still unclear whether long-term exposure to GnRHa is associated with impaired reproductive function in adulthood. The present study was performed on 46 women, former CPP patients, 12.5+/-3.7 years after the discontinuation of treatment with depot GnRHa. In a structured interview, we assessed general health status, clinical signs possibly associated with hyperandrogenism, menstrual cycle, gynaecological diseases and reproductive function. It appears that long-term treatment with depot GnRHa is safe and does not impair reproductive function. The risk of former CPP patients to develop hirsutism and/or polycystic ovary syndrome does not seem to be increased compared to the normal population but this issue needs to be addressed in further long-term follow-up studies.


Subject(s)
Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/therapeutic use , Puberty, Precocious/drug therapy , Reproduction/drug effects , Adult , Androgens/adverse effects , Body Height/drug effects , Body Mass Index , Body Weight/drug effects , Delayed-Action Preparations/administration & dosage , Drug Administration Routes , Female , Fertility/drug effects , Follow-Up Studies , Genital Diseases, Female/etiology , Gonadotropin-Releasing Hormone/administration & dosage , Health Status , Humans , Hyperandrogenism/diagnosis , Interviews as Topic , Long-Term Care , Menstrual Cycle/drug effects , Puberty, Precocious/complications , Triptorelin Pamoate/therapeutic use
14.
J Clin Endocrinol Metab ; 91(7): 2682-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16670167

ABSTRACT

CONTEXT: Congenital adrenal hyperplasia is a group of autosomal recessive inherited disorders of steroidogenesis. The deficiency of steroid 11-hydroxylase (CYP11B1) resulting from mutations in the CYP11B1 gene is the second most frequent cause. OBJECTIVE: We studied the functional and structural consequences of two CYP11B1 missense mutations, which were detected in a 1.8-yr-old boy with acne and precocious pseudopuberty, to prove their clinical relevance and study their impact on CYP11B1 function. RESULTS: The in vitro expression studies in COS-7 cells revealed an almost complete absence of CYP11B1 activity for the P94L mutant to 0.05% for the conversion of 11-deoxycortisol to cortisol. The A368D mutant severely reduced the CYP11B1 enzymatic activity to 1.17%. Intracellular localization studies by immunofluorescence revealed that the mutants were correctly localized. Introducing these mutations in a three-dimensional model structure of the CYP11B1 protein provides a possible explanation for the effects measured in vitro. We hypothesize that the A368D mutation interferes with structures important for substrate specificity and heme iron binding, thus explaining its major functional impact. However, according to structural analysis, we would expect only a minor effect of the P94L mutant on 11-hydroxylase activity, which contrasts with the observed major effect of this mutation both in vitro and in vivo. CONCLUSION: Analyzing the in vitro enzyme function is a complementary procedure to genotyping and a valuable tool for understanding the clinical phenotype of 11-hydroxylase deficiency. This is the basis for accurate genetic counseling, prenatal diagnosis, and treatment. Moreover, the combination of in vitro enzyme function and molecular modeling provides valuable insights in cytochrome P450 structural-functional relationships, although one must be aware of the limitations of in silico-based methods.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Mutation, Missense , Steroid 11-beta-Hydroxylase/genetics , Acne Vulgaris/genetics , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Cortodoxone/metabolism , Fluorescent Antibody Technique , Gene Expression , Humans , Hydrocortisone/metabolism , Infant , Male , Models, Molecular , Mutagenesis, Site-Directed , Pedigree , Puberty, Precocious/genetics , Sequence Analysis, DNA , Steroid 11-beta-Hydroxylase/chemistry , Steroid 11-beta-Hydroxylase/metabolism , Structure-Activity Relationship , Transfection
15.
Brain Dev ; 28(5): 300-4, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16478652

ABSTRACT

Satoyoshi syndrome is a rare multisystemic disorder with assumed autoimmune pathogenesis. Typical clinical features are progressive painful muscle spasms, alopecia, diarrhoea, and skeletal and endocrine abnormalities often resulting in early invalidism and death. Patients have been treated with immunoglobulins and glucocorticoids with varying outcome. We report on a 19-year-old German adolescent who has been successfully treated with a new combination of carbamazepine to reduce the severity and frequency of painful nocturnal muscle spasms, prednisolone, methotrexate and sex-steroids. Prednisolone treatment alone was not successful. After introduction of low-dose of methotrexate to the therapy the patient recovered from muscle spasms, alopecia and diarrhoea. Initiation of sex-steroid treatment resulted in pubertal development, regular menstrual cycles and improved quality of life.


Subject(s)
Endocrine System Diseases/complications , Endocrine System Diseases/drug therapy , Spasm/complications , Spasm/drug therapy , Adult , Bone Diseases, Developmental/complications , Bone Diseases, Developmental/drug therapy , Carbamazepine/therapeutic use , Drug Therapy, Combination , Female , Glucocorticoids/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Prednisolone/therapeutic use , Steroids/therapeutic use
17.
Eur J Endocrinol ; 152(4): 515-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15817905

ABSTRACT

OBJECTIVE: To clarify the molecular defect for the clinical finding of congenital hypothyroidism combined with the manifestation of calcinosis cutis in infancy. CASE REPORT: The male patient presented with moderately elevated blood thyrotropin levels at neonatal screening combined with slightly decreased plasma thyroxine and tri-iodothyronine concentrations, necessitating thyroid hormone substitution 2 weeks after birth. At the age of 7 months calcinosis cutis was seen and the patient underwent further investigation. Typical features of Albright's hereditary osteodystrophy (AHO), including round face, obesity and delayed psychomotor development, were found. METHODS AND RESULTS: Laboratory investigation revealed a resistance to parathyroid hormone (PTH) with highly elevated PTH levels and a reduction in adenylyl cyclase-stimulating protein (Gsalpha) activity leading to the diagnosis of pseudohypoparathyroidism type Ia (PHP Ia). A novel heterozygous mutation (c364T > G in exon 5, leading to the amino acid substitution Ile-106 --> Ser) was detected in the GNAS gene of the patient. This mutation was not found in the patient's parents, both of whom showed normal Gsalpha protein activity in erythrocytes and no features of AHO. A de novo mutation is therefore likely. CONCLUSIONS: Subcutaneous calcifications in infancy should prompt the clinician to a thorough search for an underlying disease. The possibility of AHO and PHP Ia should be considered in children with hypothyroidism and calcinosis cutis. Systematic reviews regarding the frequency of calcinosis in AHO are warranted.


Subject(s)
Calcinosis/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Pseudohypoparathyroidism/genetics , Calcinosis/complications , Chromogranins , Diagnosis, Differential , Fibrous Dysplasia, Polyostotic , Heterozygote , Humans , Hypothyroidism/complications , Infant, Newborn , Male , Pseudohypoparathyroidism/complications , Pseudohypoparathyroidism/diagnosis
18.
J Mol Med (Berl) ; 83(7): 561-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15830218

ABSTRACT

Congenital adrenal hyperplasia (CAH) [OMIM 201910] is a group of autosomal recessive disorders, caused in 90-95% of cases by a deficiency of steroid 21-hydroxylase due to mutations in the CYP21A2 gene. The functional and structural effects of a novel rare missense mutation (E351K) in CYP21A2 found in a male patient with simple virilizing CAH were studied. The novel E351K point mutation is located in the ERR triad of the 21-hydroxylase. The ERR triad is a glutamine-arginine-arginine motif conserved in all cytochrome P450 sequences. The glutamate and first arginine residue are invariant in all P450 cytochrome enzymes, whereas the second arginine residue is present as arginine, histidine, or asparagine. Although the ERR triad is involved in some way to heme binding by the cytochrome P450 monooxygenases, the E351K mutation leads to severe but not complete loss of CYP21 enzyme activity. The functional analysis in COS-7 cells revealed a reduced conversion of 17-hydroxyprogesterone to 11-deoxycortisol of 1.1+/-0.5% (SD) and of progesterone to 11-deoxycorticosterone of 1.2+/-0.3% of wild-type activity. Analyzing the artificial mutants (E351D, E351I) of the E351 residue did not show a restoration of the in vitro 21-hydroxylase activity. These effects could be readily explained by structural changes induced by the mutations, which were rationalized by a three-dimensional-model structure of the CYP21 protein. The combination of in vitro enzyme function and computerized protein analysis of the E351 residue of the CYP21 protein provides experimental evidence for the ERR triad being a fundamental structural element of cytochrome P450 enzymes.


Subject(s)
Amino Acid Substitution/genetics , Point Mutation/genetics , Steroid 21-Hydroxylase/chemistry , Steroid 21-Hydroxylase/metabolism , Animals , COS Cells , Child, Preschool , Chlorocebus aethiops , Humans , Infant, Newborn , Male , Models, Molecular , Pedigree , Protein Transport , Sequence Analysis, DNA , Steroid 21-Hydroxylase/genetics , Structure-Activity Relationship
19.
Hum Mutat ; 25(5): 502-3, 2005 May.
Article in English | MEDLINE | ID: mdl-15841486

ABSTRACT

X-linked adrenal hypoplasia congenita (AHC) is a rare developmental disorder associated with primary adrenal insufficiency and combined primary and secondary male hypogonadism. It is caused by deletions or mutations of the NR0B1 (DAX1) gene encoding DAX1, an atypical orphan member of the nuclear receptor superfamily. The continuous molecular genetic analysis of male patients with primary adrenal insufficiency revealed 13 novel mutations within the coding region of the NR0B1 gene which are predicted to inactivate the DAX1 function. These were three nonsense mutations (c.312C>A, p.Cys104X, c.670C>T, p.Gln224X; and c.873G>A, p.Trp291X), five duplications (c.269_270dup, c.421_422dup, c.895_896dup, c.989dup, c.999_1000dup), and five deletions (c.483del, c.745_746del, c.734_740del, c.1092del, and c.1346del). All of the mutations resulted in a premature stop codon destroying the ligand binding domain of the predictive DAX1 protein.


Subject(s)
Adrenal Insufficiency/genetics , DNA-Binding Proteins/genetics , Mutation , Receptors, Retinoic Acid/genetics , Repressor Proteins/genetics , Child , Child, Preschool , Codon, Nonsense , DAX-1 Orphan Nuclear Receptor , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , Gene Deletion , Gene Duplication , Humans , Infant , Infant, Newborn , Male , Protein Structure, Tertiary/genetics , Receptors, Retinoic Acid/chemistry , Repressor Proteins/chemistry
20.
J Clin Endocrinol Metab ; 90(6): 3724-30, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15755848

ABSTRACT

Congenital adrenal hyperplasia is a group of autosomal recessive disorders second most often caused by deficiency of steroid 11-hydroxylase (CYP11B1) due to mutations in the CYP11B1 gene. We studied the functional and structural consequences of two novel missense mutations (W116C, L299P) and an in-frame 3-bp deletion (DeltaF438) in the CYP11B gene, detected in three unrelated families. All patients are suffering from classical CYP11B1 deficiency. In vitro expression studies in COS-7 cells revealed a decreased CYP11B1 activity in the W116C mutant to 2.9 +/- 0.9% (sd) for the conversion of 11-deoxycortisol to cortisol. The L299P mutant reduced the enzymatic activity to 1.2 +/- 0.9%, whereas the DeltaF438 mutation resulted in no measurable residual CYP11B1 activity. Introduction of these mutations in a three-dimensional model structure of the CYP11B1 protein provides a possible explanation for the in vitro measured effects. We hypothesize that the W116C mutation influences the conformational change of the 11-hydroxylase protein necessary for substrate access and product release. The L299P mutation causes a change in the position of the I helix relative to the heme group, whereas the DeltaF438 mutation results in a steric disarrangement of the heme group relative to the enzyme. Studying the enzyme function in vitro helps to understand the phenotypical expression and disease severity of 11-hydroxylase deficiency, which is the basis for accurate genetic counseling, prenatal diagnosis, and treatment. Moreover, the combination of in vitro enzyme function and molecular modeling provides new insights in cytochrome P450 structural-functional relationships.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Base Pairing , Point Mutation , Sequence Deletion , Steroid 11-beta-Hydroxylase/genetics , Adolescent , Adrenal Cortex Hormones/blood , Adrenal Hyperplasia, Congenital/enzymology , Adrenocorticotropic Hormone , Amino Acid Substitution , Child , Female , Genes, Recessive , Genitalia, Female/abnormalities , Humans , Kinetics , Male , Models, Molecular , Pedigree , Protein Conformation , Steroid 11-beta-Hydroxylase/chemistry , Steroid 11-beta-Hydroxylase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...