Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Methods Mol Biol ; 776: 3-24, 2011.
Article in English | MEDLINE | ID: mdl-21796517

ABSTRACT

Androgens and the androgen receptor (AR) are indispensable for expression of the male phenotype. The two most important androgens are testosterone and 5α-dihydrotestosterone. The elucidation of the mechanism of androgen action has a long history starting in the 19th century with the classical experiments by Brown-Séquard. In the 1960s the steroid hormone receptor concept was established and the AR was identified as a protein entity with a high affinity and specificity for testosterone and 5α-dihydrotestosterone. In addition, the enzyme 5α-reductase type 2 was discovered and found to catalyze the conversion of testosterone to the more active metabolite 5α-dihydrotestosterone. In the second half of the 1980s, the cDNA cloning of all steroid hormone receptors, including that of the AR, has been another milestone in the whole field of steroid hormone action. Despite two different ligands (testosterone and 5α-dihydrotestosterone), only one AR cDNA has been identified and cloned. The AR (NR3C4) is a ligand-dependent transcription factor and belongs to the family of nuclear hormone receptors which has 48 members in human. The current model for androgen action involves a multistep mechanism. Studies have provided insight into AR association with co-regulators involved in transcription initiation and on intramolecular interactions of the AR protein during activation. Knowledge about androgen action in the normal physiology and in disease states has increased tremendously after cloning of the AR cDNA. Several diseases, such as androgen insensitivity syndrome (AIS), prostate cancer and spinal bulbar muscular atrophy (SBMA), have been shown to be associated with alterations in AR function due to mutations in the AR gene or dysregulation of androgen signalling. A historical overview of androgen action and salient features of AR function in normal and disease states are provided herein.


Subject(s)
Androgens/genetics , Androgens/metabolism , Androgens/history , Androgens/physiology , Animals , History, 19th Century , History, 20th Century , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Testosterone/biosynthesis , Testosterone/metabolism
2.
Biochim Biophys Acta ; 1794(2): 193-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19059367

ABSTRACT

An oligonucleotide-based assay (OBA) was used to identify novel co-factors that can be recruited by the deoxyribonucleic acid (DNA)-bound androgen receptor (AR). Nuclear extracts obtained from LNCaP cells, after incubation with R1881, were incubated with biotinylated oligonucleotides bound to streptavidin coated beads. The oligonucleotides contain 3 copies in tandem of the androgen responsive element ARE1 from the prostate specific antigen (PSA) gene promoter. As control incubation, a scrambled version of the tandem ARE1 was used. Immunoblots of the eluents revealed that the AR was bound to the ARE1 oligonucleotide and to a much lesser extent to the scrambled oligonucleotide. Proteins eluted from the oligonucleotides, were separated on a 5-15% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gradient gel, followed by identification using mass spectrometry. Identified proteins were scored for having one or more of the following known properties: nuclear localization, involved in transcription regulation, involvement in steroid hormone receptor (SHR) function, or specifical involvement in AR function. A total number of 85 nuclear proteins were found in two separate OBAs. Based on peptide counting, we found enrichment of 7 proteins eluted from the ARE1 oligonucleotide, compared to the scrambled oligonucleotide. Taken together with the obtained scores, these proteins are considered putative AR co-factors. One of these proteins, DDX17, is known to be a co-factor for estrogen receptor alpha (ERalpha), but has never been associated with AR function. The results indicate that the ARE oligonucleotide-based assay may allow enrichment of new candidate DNA-bound AR interacting proteins.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA/metabolism , Receptors, Androgen/physiology , Biotin , Cell Line, Tumor , Humans , Male , Metribolone/pharmacology , Oligonucleotides , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Protein Binding , Protein Interaction Mapping , Receptors, Androgen/genetics , Response Elements , Streptavidin
3.
Mol Cell Endocrinol ; 292(1-2): 69-78, 2008 Sep 24.
Article in English | MEDLINE | ID: mdl-18656523

ABSTRACT

A novel mutation F826L located within the ligand binding domain (LBD) of the human androgen receptor (AR) was investigated. This mutation was found in a boy with severe penoscrotal hypospadias (classified as 46,XY DSD). The AR mutant F826L appeared to be indistinguishable from the wild-type AR, with respect to ligand binding affinity, transcriptional activation of MMTV-luciferase and ARE2-TATA-luciferase reporter genes, protein level in genital skin fibroblasts (GSFs), and sub-cellular distribution in transfected cells. However, an at least two-fold higher NH2-/COOH-terminal domain interaction was found in luciferase and GST pull-down assays. A two-fold increase was also observed for TIF2 (transcription intermediary factor 2) co-activation of the AR F826L COOH-terminal domain. This increase could not be explained by a higher stability of the mutant protein, which was within wild-type range. Repression of transactivation by the nuclear receptor co-repressor (N-CoR) was not affected by the AR F826L mutation. The observed properties of AR F826L would be in agreement with an increased activity rather than with a partial defective AR transcriptional activation. It is concluded that the penoscrotal hypospadias in the present case is caused by an as yet unknown mechanism, which still may involve the mutant AR.


Subject(s)
Amino Acid Substitution , Androgen-Insensitivity Syndrome/genetics , Mutation/genetics , Nuclear Receptor Coactivator 2/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Cell Line , Child, Preschool , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Foreskin/cytology , Humans , Immunoprecipitation , Infant , Ligands , Male , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Subcellular Fractions/metabolism , Transcriptional Activation/genetics
4.
Biochim Biophys Acta ; 1774(5): 575-82, 2007 May.
Article in English | MEDLINE | ID: mdl-17467349

ABSTRACT

The aim of the present study was to identify proteins differentially regulated by TRPS1 in human prostate cancer cells in order to better understand the role of TRPS1 in prostate cancer development. The proteomes of androgen-independent DU145 prostate cancer cells, that do not express TRPS1 and of genetically engineered DU145 cells that stable and inducible express recombinant TRPS1 protein, were compared. Using two-dimensional electrophoresis followed by mass spectrometric analysis, 13 proteins that were differentially expressed between these two cell lines were identified. These proteins represent a dominant reduction of expression of antioxidant proteins, including superoxide dismutase, protein disulfide isomerase A3 precursor, endoplasmin precursor and annexin A2. Furthermore, regulation was observed for mitochondrion-associated proteins, glycolytic enzymes, a cytoskeleton-associated protein, a nuclear protein and proteins involved in apoptosis. Our data indicate that overexpression of TRPS1 protein is correlated with reduced protein expression of certain antioxidants. This suggests a possible involvement of TRPS1 in oxidative stress, and possibly in apoptosis in androgen-independent DU145 prostate cancer cells.


Subject(s)
DNA-Binding Proteins/physiology , Prostatic Neoplasms/metabolism , Proteome , Transcription Factors/physiology , Amino Acid Sequence , Antioxidants/metabolism , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Male , Mass Spectrometry , Molecular Sequence Data , Prostatic Neoplasms/pathology , Repressor Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism
5.
J Mol Endocrinol ; 38(1-2): 91-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17242172

ABSTRACT

The glucocorticoid receptor (GR) is widely expressed in various tissues throughout the human body. At least three different 3'-splice variants of the GR have been reported: GR-alpha, which is functionally active; GR-beta, which is a dominant negative inhibitor of GR-alpha function; and GR-P, which is thought to activate the function of GR-alpha. At least seven different variants for exon 1 exist, 1A-1F and 1H, each with its own promoter. In this study, we explored if tissue-specific splicing of the 3'-end variants of the GR is influenced by alternative promoter usage. cDNAs of different tissues and cell lines were used to investigate which part of transcripts carrying each of the three major variants for exons 1, 1A, 1B, or 1C, encodes for the splice variants GR-alpha, GR-beta, and GR-P. Our data demonstrate that the expression of GR-alpha is preferentially regulated by promoter 1C and that for the expression of GR-P promoter 1B is predominantly used. This indicates that regulation of GR splice variants could partly occur through selective use of the multiple promoters, and that this is another way to sensitize cells and tissues to the different activities of the GR isoforms.


Subject(s)
Alternative Splicing/genetics , Promoter Regions, Genetic , Receptors, Glucocorticoid/genetics , Cell Line, Transformed , Exons , Gene Expression Regulation/physiology , Humans , Organ Specificity , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Receptors, Glucocorticoid/biosynthesis
6.
J Clin Endocrinol Metab ; 92(1): 359-62, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17062770

ABSTRACT

CONTEXT: Recently, it was proposed that a combination of the 83,557insA polymorphism in the 11beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) gene and the R453Q polymorphism in the hexose-6-phosphate dehydrogenase (H6PD) gene interacts to cause cortisone reductase deficiency (CRD) when at least three alleles are affected. OBJECTIVE: The aim was to study the separate and combined effects of these polymorphisms on body composition, adrenal androgen production, blood pressure, glucose metabolism, and the incidence of dementia in the healthy elderly population. DESIGN/SETTING/PARTICIPANTS: The Rotterdam study (n = 6105) and the Frail Old Men study (n = 347) are population-based cohort studies in the elderly. MAIN OUTCOME MEASURES: Genotype distributions and influences of (combined) genotypes on body mass index, adrenal androgen production, waist to hip ratio, systolic and diastolic blood pressure, fasting glucose levels, glucose tolerance test, and incidence of dementia were measured. RESULTS: No influence of the HSD11B1 83,557insA (allele frequencies 22.0 and 21.5%) and H6PD R453Q (allele frequencies 22.9 and 20.2%) variants was found for the different outcome measures that were investigated, either separately or when at least three alleles were affected. CONCLUSIONS: Two population-based studies among Caucasian elderly showed no evidence for (combined) effects of two polymorphisms in the HSD11B1 and H6PD genes on body composition, adrenal androgen production, blood pressure, glucose metabolism, and incidence of dementia. Moreover, the high frequencies observed for these two polymorphisms do not correspond to the low incidence of CRD observed in the general population. Altogether, it is unlikely that these polymorphisms cause CRD.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/genetics , Androgens/biosynthesis , Blood Pressure , Body Composition , Carbohydrate Dehydrogenases/genetics , Cortisone Reductase/deficiency , Dementia/genetics , Glucose/metabolism , Polymorphism, Single Nucleotide , Adrenal Glands/metabolism , Aged , Body Mass Index , Dementia/enzymology , Humans , Male , Waist-Hip Ratio
7.
J Clin Endocrinol Metab ; 91(7): 2800-3, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16684836

ABSTRACT

CONTEXT: Glucocorticoids (GCs) are extensively used in the treatment of inflammatory and autoimmune diseases. Their beneficial effects are thought to be mediated by GC transrepression on gene expression. However, their use is limited by serious adverse effects, presumably mediated by GC transactivation of gene expression. OBJECTIVE: The objective of the study was to investigate the effect of the GC receptor haplotype, characterized by the GR-9beta polymorphism, on GC transactivation and transrepression. DESIGN AND METHODS: This was a cross-sectional study in 216 persons randomly selected from participants in The Rotterdam Study, a population-based cohort study in the elderly. Clinical and biochemical parameters of GC sensitivity were measured: weight, height, waist to hip ratio, glucose, insulin, total cholesterol, high-density lipoprotein, and C-reactive protein. In a dexamethasone suppression test, the response of serum cortisol concentrations was studied. Genotyping for four GC receptor polymorphisms was performed. In addition, ex vivo experiments were performed with leukocytes of 10 healthy controls and two persons homozygous for the GR-9beta polymorphism, in which the expression of two GC-sensitive genes, GC-induced leucine zipper and IL-2, was measured. RESULTS: Persons carrying the GR-9beta haplotype without 22/23EK (n = 53) revealed no significant differences in their body mass index, waist to hip ratio, fat spectrum, and insulin sensitivity or in their cortisol response to dexamethasone and levels of C-reactive protein, compared with noncarriers (n = 113). Ex vivo, GC-induced up-regulation of GC-induced leucine zipper mRNA via transactivation did not significantly differ in GR-9beta homozygotes, whereas the down-regulation of IL-2 expression via transrepression was decreased. CONCLUSION: Persons carrying the GR-9beta haplotype seem to have a decreased GC transrepression with normal transactivation.


Subject(s)
Gene Expression Regulation/genetics , Polymorphism, Genetic/genetics , Receptors, Glucocorticoid/genetics , Aged , Aging , Body Mass Index , C-Reactive Protein/analysis , Cohort Studies , Cross-Sectional Studies , Dexamethasone , Down-Regulation/genetics , Female , Gene Expression , Genotype , Glucocorticoids/pharmacology , Heterozygote , Humans , Hydrocortisone/blood , Insulin Resistance , Interleukin-2/genetics , Leucine Zippers/genetics , Male , Transcriptional Activation/genetics , Waist-Hip Ratio
8.
Mol Endocrinol ; 20(8): 1742-55, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16627595

ABSTRACT

The androgen receptor (AR) ligand-binding domain (LBD) binds FXXLF motifs, present in the AR N-terminal domain and AR-specific cofactors, and some LXXLL motifs of nuclear receptor coactivators. We demonstrated that in the context of the AR FXXLF motif many different amino acid residues at positions +2 and +3 are compatible with strong AR LBD interaction, although a preference for E at +2 and K or R at +3 was found. Pairwise systematic analysis of F/L swaps at +1 and +5 in FXXLF and LXXLL motifs showed: 1) F to L substitutions in natural FXXLF motifs abolished AR LBD interaction; 2) binding of interacting LXXLL motifs was unchanged or increased upon L to F substitutions; 3) certain noninteracting LXXLL motifs became strongly AR-interacting FXXLF motifs; whereas 4) other nonbinders remained unaffected by L to F substitutions. All FXXLF motifs, but not the corresponding LXXLL motifs, displayed a strong preference for AR LBD. Progesterone receptor LBD interacted with some FXXLF motifs, albeit always less efficiently than corresponding LXXLL motifs. AR LBD interaction of most FXXLF and LXXLL peptides depended on classical charge clamp residue K720, whereas E897 was less important. Other charged residues lining the AR coactivator-binding groove, K717 and R726, modulated optimal peptide binding. Interestingly, these four charged residues affected binding of individual peptides independent of an F or L at +1 and +5 in swap experiments. In conclusion, F residues determine strong and selective peptide interactions with AR. Sequences flanking the core motif determine the specific mode of FXXLF and LXXLL interactions.


Subject(s)
Leucine Zippers/physiology , Leucine/metabolism , Phenylalanine/metabolism , Protein Interaction Mapping , Receptors, Androgen/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Amino Acid Motifs/physiology , Binding Sites , Cells, Cultured , DNA Mutational Analysis , Dimerization , Histone Acetyltransferases , Humans , Ligands , Models, Molecular , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2/metabolism , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Protons , Receptors, Androgen/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Homology, Amino Acid , Transcription Factors/metabolism , Transfection , Two-Hybrid System Techniques
9.
J Clin Endocrinol Metab ; 91(2): 694-701, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16317053

ABSTRACT

CONTEXT: The clinical presentation of abnormalities in glucocorticoid (GC) sensitivity is diverse, and therefore it is difficult to diagnose this condition. OBJECTIVE AND DESIGN: The objective of the study was to develop strategies for the characterization of GC sensitivity disorders. SETTING: The study was conducted in an outpatient clinic. PATIENTS: Nine patients with GC sensitivity disorders participated. INTERVENTIONS: Sequence analysis of the GC receptor (GR), determination of GR number per cell, GR ligand-binding affinity, and GR splice regulation were performed in freshly prepared peripheral blood mononuclear lymphocytes and Epstein-Barr virus-transformed lymphoblasts. Cellular GC sensitivity was determined ex vivo by measuring the effect of dexamethasone on GC-induced leucine-zipper and IL-2 mRNA levels and on cell proliferation. RESULTS: Differences in GR number per cell, GR affinity, GR splice variants, and effects on transactivation or transrepression of GC-sensitive genes were observed between patients and controls. Epstein-Barr virus transformation of lymphoblasts had no influence on GR affinity but increased the GR number 5-fold in healthy controls. In patients diagnosed as cortisol resistant, however, GR number after transformation was increased significantly less than 5-fold, whereas a higher GR number was observed in a patient suspected of cortisol hypersensitivity. CONCLUSION: This study illustrates several strategies to define abnormalities in GC sensitivity by describing nine patients with affected GC sensitivity, all with a unique clinical course and background.


Subject(s)
Adrenal Cortex Diseases/metabolism , Glucocorticoids/metabolism , Hydrocortisone/metabolism , Receptors, Glucocorticoid/metabolism , Adolescent , Adrenal Cortex Diseases/genetics , Adrenal Cortex Diseases/pathology , Adult , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line, Transformed , Cell Transformation, Viral , DNA/chemistry , DNA/genetics , Female , Gene Dosage , Humans , Male , Polymorphism, Single Nucleotide , Receptors, Glucocorticoid/genetics , Sequence Analysis, DNA
10.
J Clin Endocrinol Metab ; 90(11): 6237-43, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16118334

ABSTRACT

CONTEXT: The effects of cortisol are mediated by the alpha-isoform of the glucocorticoid receptor (GR). GR-alpha levels and activity are modulated by alternative splicing of the common pre-mRNA into mRNAs for the GR-beta and GR-P isoforms. OBJECTIVE: The objective of this study was to investigate whether chronic hypercortisolism, chronic hypocortisolism, or acute, relative hypocortisolism influences the expression levels of the GR splice variants in mononuclear leukocytes. DESIGN: This was a case-control study. SETTING: The study was performed at a university hospital. PARTICIPANTS: Eighteen patients with Cushing's syndrome, five patients with hypocortisolemia, seven patients undergoing metyrapone testing, and 14 controls were studied. MAIN OUTCOME MEASURES: The main outcome measures were mRNA levels, GR affinity, and number per cell. RESULTS: All three GR mRNA isoforms were detected in participants from all groups at relative levels of alpha/P/beta = 1:0.25:0.001. There was a significant correlation between the expression levels of the three splice variants and between the mRNA levels and the number of receptors per cell. The GR in Cushing patients had an increased Kd (P < 0.05) preoperatively. GR number was not significantly different. Postoperatively, the Kd decreased. GR-beta mRNA expression was increased compared with controls (P < 0.05) and was decreased after surgery (P < 0.05). In patients with chronic hypocortisolism, GR-alpha mRNA expression was increased, and receptor numbers were increased (P < 0.05), whereas GR affinity was normal. No changes were observed in patients undergoing a metyrapone test. CONCLUSIONS: Cushing's syndrome is accompanied by a reversible decrease in GR affinity, possibly related to an increased GR-beta expression, which may be a compensatory mechanism to GC excess. In chronic hypocortisolism, adaptive changes in GR status seem to occur at the level of GR number.


Subject(s)
Cushing Syndrome/metabolism , Hydrocortisone/deficiency , Leukocytes, Mononuclear/chemistry , Receptors, Glucocorticoid/blood , Adult , Aged , Case-Control Studies , Female , Humans , Hydrocortisone/blood , Male , Metyrapone , Middle Aged , Protein Isoforms , RNA, Messenger/analysis , Receptors, Glucocorticoid/genetics
11.
J Clin Endocrinol Metab ; 90(10): 5804-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16030164

ABSTRACT

CONTEXT: Interindividual variation in glucocorticoid (GC)-sensitivity can be partly explained by polymorphisms in the GC receptor (GR) gene. The ER22/23EK and N363S polymorphisms have been described to be associated with lower and higher GC sensitivity, respectively. OBJECTIVE AND DESIGN: We examined the basis of this altered GC sensitivity by expressing GR(N363S) and GR(ER22/23EK) in COS-1 cells and investigating their transactivating and transrepressing capacities using a GC response element-luciferase reporter and a p65-activated nuclear factor kappaB-luciferase reporter, respectively. Furthermore, we evaluated the transactivating and transrepressing capacities of the GR in peripheral blood mononuclear lymphocytes of homozygous and heterozygous carriers of these polymorphisms by determining the maximum effect of dexamethasone on transactivation of the GC-induced leucine-zipper and transinhibition of the IL-2 gene by means of real-time RT-PCR. RESULTS: The effects of the polymorphisms in the GR gene previously observed in population studies were also detected at the level of gene expression. The ER22/23EK polymorphism resulted in a significant reduction of transactivating capacity, in both transfection experiments (-14 +/- 5%, P < 0.05) and peripheral blood mononuclear lymphocytes of carriers of this polymorphism (homozygous: -48 +/- 6%, P < 0.01, n = 1; heterozygous: -21 +/- 4%, P = 0.08, n = 3). The N363S polymorphism, associated with increased GC sensitivity, resulted in a significantly increased transactivating capacity, both in vitro (8 +/- 3%; P < 0.02) and ex vivo (homozygous: 204 +/- 19%, P < 0.0001, n = 1; heterozygous: 124 +/- 8%, P = 0.05, n = 3). Neither the ER22/23EK nor the N363S polymorphism seemed to influence the transrepressing capacity of the GR. CONCLUSION: The presence of these and other GC sensitivity-modulating polymorphisms may have consequences for the use of GCs in a clinical setting.


Subject(s)
Gene Expression Regulation/physiology , Polymorphism, Genetic/genetics , Receptors, Glucocorticoid/genetics , Animals , Blotting, Western , COS Cells , Cells, Cultured , Chlorocebus aethiops , DNA Primers/genetics , Dexamethasone/pharmacology , Genetic Variation , Interleukin-2/biosynthesis , Interleukin-2/genetics , Luciferases/genetics , Plasmids/genetics , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
12.
J Clin Endocrinol Metab ; 90(9): 5313-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15985487

ABSTRACT

CONTEXT: CYP3A7, expressed in the human fetal liver and normally silenced after birth, plays a major role in the 16alpha-hydroxylation of dehydroepiandrosterone (DHEA), DHEA sulfate (DHEAS), and estrone. Due to a replacement of part of the CYP3A7 promoter with a sequence identical with the same region in the CYP3A4 promoter (referred to as CYP3A7*1C), some individuals still express a variant of the CYP3A7 gene later in life. OBJECTIVE: The objective of this study was to examine the effect of the CYP3A7*1C polymorphism on serum steroid hormone levels. DESIGN, SETTING, PARTICIPANTS: Two population-based cohort studies were performed. Study group 1 consisted of 208 subjects randomly selected from the Rotterdam Study, and study group 2 consisted of 345 elderly independently living men. MAIN OUTCOME MEASURES: Serum DHEA(S), androstenedione, estradiol, estrone, and testosterone levels were the main outcome measures. RESULTS: In study groups 1 and 2, heterozygous CYP3A7*1C carriers had almost 50% lower DHEAS levels compared with homozygous carriers of the reference allele [study group 1, 1.74 +/- 0.25 vs. 3.33 +/- 0.15 micromol/liter (P = 0.02); study group 2, 2.09 +/- 0.08 vs. 1.08 +/- 0.12 micromol/liter (P < 0.001)]. No differences in circulating DHEA, androstenedione, estradiol, or testosterone levels were found. However, in study group 2, serum estrone levels were lower in heterozygous CYP3A7*1C carriers compared with homozygous carriers of the reference allele (0.11 +/- 0.002 vs. 0.08 +/- 0.006 nmol/liter; P < 0.001). CONCLUSION: The CYP3A7*1C polymorphism causes the persistence of enzymatic activity of CYP3A7 during adult life, resulting in lower circulating DHEAS and estrone levels.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Dehydroepiandrosterone Sulfate/blood , Polymorphism, Genetic , Aged , Aged, 80 and over , Androstenedione/blood , Cohort Studies , Cytochrome P-450 CYP3A , Estradiol/blood , Estrone/blood , Genotype , Heterozygote , Homozygote , Humans , Male , Middle Aged , Testosterone/blood
13.
J Soc Gynecol Investig ; 12(4): 285-92, 2005 May.
Article in English | MEDLINE | ID: mdl-15866122

ABSTRACT

OBJECTIVE: Progesterone plays an important role in controlling proliferation and differentiation of the human endometrium. Because there are two progesterone receptor isoforms (PRA and PRB), it was important to generate tools to be able to study the role of these two progesterone receptors separately. METHODS: Using stable transfection techniques, both human progesterone receptor isoforms (hPRA and hPRB) were reintroduced into a hPR-negative subclone of the well-differentiated endometrial cancer cell line Ishikawa. Several Ishikawa subcell lines were constructed, each expressing different levels of hPRA, hPRB, or hPRA and hPRB, respectively. RESULTS: These Ishikawa subcell lines showed a marked progesterone-induced growth inhibition with induction of apoptosis after long-term culture in the presence of hormone. Upon measuring gene regulation, a clear difference in regulation of expression of the selected genes by progesterone treatment was observed between the PRA-, PRB-, or PRA/B-expressing cell lines. Integrin beta4 (ITGB4) was only regulated in PRA-expressing cells; amphiregulin was highly regulated in PRB-expressing cells; insulin-like growth factor binding protein 3 (IGFBP3) was only regulated in PRB- and PRA/B-expressing cells; and metallothionein 1L (MT1L) was highly regulated in PRA/B-expressing cells. Interestingly, based on literature data, these genes can be implicated in induction of apoptosis, but are modulated here in such a way that suggests induction of resistance against apoptosis. CONCLUSION: Reintroduction of PRs into Ishikawa cells rescued progesterone responsiveness in these cells. Furthermore, using these human endometrial cancer subcell lines, clear and distinct functional differences between the PR isoforms were observed.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Progesterone/physiology , Receptors, Progesterone/biosynthesis , Apoptosis , Cell Differentiation , Cell Proliferation , Female , Gene Expression Profiling , Humans , Protein Isoforms , Transfection , Tumor Cells, Cultured
14.
Mol Endocrinol ; 19(7): 1687-96, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15746190

ABSTRACT

One of the most intriguing polymorphisms in the GR [glucocorticoid (GC) receptor] gene is in codons 22 and 23 [GAGAGG(GluArg) --> GAAAAG (GluLys)]. This polymorphism is associated with a reduced GC sensitivity, a better metabolic and cardiovascular health profile, and an increased survival rate. Recently, Yudt and Cidlowski reported that two different methionine codons in the GR mRNA may be used as initiation codon: AUG-1 and AUG-27, resulting in two isoforms, the GR-A and the GR-B proteins, respectively. They also showed that the GR-B protein had a stronger transactivating effect in transient transfection experiments. In this study, we elucidated the molecular basis for the reduced GC sensitivity by investigating the influence of the ER22/23EK polymorphism on synthesis of GR-A and GR-B by expressing them independently from constructs with and without the polymorphic site. Binding studies with [(3)H]-dexamethasone and transactivation studies showed that, when the ER22/23EK polymorphism is present, approximately 15% more GR-A protein was expressed, whereas total GR levels (GR-A + GR-B) were not affected. These results show that the transcriptional activity in GR(ER22/23EK) carriers is decreased because more of the less transcriptionally active GR-A isoform is formed. This is probably caused by altered secondary mRNA structure.


Subject(s)
Polymorphism, Genetic , Receptors, Glucocorticoid/genetics , Alternative Splicing/genetics , Animals , Arginine/genetics , Base Sequence , COS Cells , Chlorocebus aethiops , Codon, Initiator/genetics , Dexamethasone/metabolism , Humans , Lysine/genetics , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Peptide Chain Initiation, Translational , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Transcription, Genetic , Transcriptional Activation/genetics
15.
J Clin Endocrinol Metab ; 90(5): 2994-3000, 2005 May.
Article in English | MEDLINE | ID: mdl-15728202

ABSTRACT

Individual glucocorticoid (GC) sensitivity was determined by measuring the effects of several clinically used GCs on transactivation of the GC-induced leucine zipper (GILZ) gene and on transrepression of the IL-2 gene using quantitative real-time PCR. A clear difference in relative potencies for transactivation and transrepression of the various GCs was observed, suggesting differential effects. To determine whether the in vitro outcomes could predict in vivo effects of GCs, 15 individuals underwent a 0.25-mg dexamethasone (DEX) suppression test (DST) while determining GILZ and IL-2 mRNA levels in their peripheral blood mononuclear cells incubated with hydrocortisone, DEX, budesonide, and prednisolone. No correlations were found between the DST and the two expression assays. However, significant correlations existed between hydrocortisone and DEX (r = 0.52; P = 0.046), hydrocortisone and budesonide (r = 0.48; P = 0.069), and hydrocortisone and prednisolone (r = 0.86; P = 0.007) regarding GILZ mRNA levels, and between hydrocortisone and DEX (r = 0.62; P = 0.014), hydrocortisone and budesonide (r = 0.71; P = 0.003), and hydrocortisone and prednisolone (r = 0.71; P = 0.047) regarding IL-2 mRNA levels. In conclusion, intra- and inter-individual variations in GC sensitivity were observed using two expression assays representing GC-mediated transactivation and transrepression. The two expression assays did not correlate with each other or with the results of the DST. This suggests that regulation of the hypothalamic-pituitary-adrenal axis is more complex. However, within an individual person, these two tests combined might predict what type and dosage of GC will be preferable in individual patients for its inhibitory clinical effects, together with relatively fewer transactivating effects related to adverse effects.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Interleukin-2/genetics , Transcription Factors/genetics , Dexamethasone/pharmacology , Humans , Hydrocortisone/blood , Male , Middle Aged , Receptors, Glucocorticoid/physiology , Transcriptional Activation
16.
J Clin Endocrinol Metab ; 90(1): 507-15, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15486055

ABSTRACT

Androgen insensitivity syndrome (AIS) is caused by defects in the androgen receptor (AR) that render the AR partially or completely inactive. As a result, embryonic sex differentiation is impaired. Here, we describe a novel mutation in the AR found in a patient with partial AIS. The mutation results in a substitution of a glutamine (Q) by a lysine (K) residue at position 902, Q902K. The AR Q902K mutation was investigated in vitro with respect to its functional properties. The equilibrium dissociation constants (K(d)s) of AR Q902K in the presence of either the synthetic androgen R1881 or the natural ligand DHT were slightly elevated. The R1881 dissociation rate (t(1/2)) was increased 3-fold for AR Q902K compared with wild type. Transcriptional activity was decreased to 85% of wild type, and the dose-response curve revealed that the sensitivity to hormone was decreased due to the mutation. Furthermore, the 114-kDa androgen-induced phosphorylated AR protein band was not detectable in genital skin fibroblasts. However, it could be detected in transfected CHO cells expressing the mutant receptor in the presence of 10 and 100 nm R1881. Functional interaction assays and a GST pull-down assay showed that the interaction between the NH2 and COOH terminus of AR Q902K was reduced to 50% of wild type. Furthermore, the transactivation by the coactivator TIF2 (transcriptional intermediary factor 2) was decreased 2- to 3-fold. The half-maximal response in both assays was shifted to a higher hormone concentration compared with wild type. These results indicate that residue Q902 is involved in TIF2 and NH2/COOH interaction and that the Q to K mutation results in a mild impairment of AR function, which can explain the partial AIS phenotype of the patient.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Mutation , Receptors, Androgen/genetics , Animals , CHO Cells , Child, Preschool , Cricetinae , Humans , Male , Receptors, Androgen/chemistry , Receptors, Androgen/physiology , Transcriptional Activation
17.
J Biol Chem ; 280(9): 7786-92, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15623502

ABSTRACT

Androgen receptor (AR) induced precocious myogenesis in culture and myogenic specified gene activity. Increased levels of AR expression in replicating C2C12 myoblasts stimulated fusion into post-differentiated multinucleated myotubes and the appearance of skeletal alpha-actin transcripts, even in the absence of ligand. Furthermore, AR activated the skeletal alpha-actin promoter, which lacks GRE sites, in co-transfected C2C12 cells. AR co-activation of the skeletal alpha-actin promoter required co-expressed full-length serum response factor (SRF). In vitro, AR associated with SRF and was recruited by SRF to a alpha-actin promoter SRF binding site. Our data suggest that AR is capable of activating myogenic genes devoid of consensus AR binding sites via its recruitment by the myogenic enriched transcription factor, SRF.


Subject(s)
Receptors, Androgen/chemistry , Serum Response Factor/metabolism , Actins/metabolism , Animals , Binding Sites , Cell Differentiation , Cell Line , Cells, Cultured , DNA/metabolism , DNA Primers/chemistry , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Ligands , Mice , Muscle, Skeletal/metabolism , Muscles/cytology , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Time Factors , Transcription, Genetic , Transfection
18.
Endocr Relat Cancer ; 11(4): 815-22, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15613454

ABSTRACT

TRPS1 mRNA is more highly expressed in androgen-dependent lymph node carcinoma of prostate-fast growing colony (LNCaP-FGC) compared with androgen-independent lymph node carcinoma of prostate-lymph node original (LNCaP-LNO) prostate cancer cell lines. Furthermore, TRPS1 mRNA expression is down-regulated by androgens in LNCaP-FGC cells, a process mediated by the androgen receptor (AR). Here, we present TRPS1 protein expression in human prostate cancer material derived from a panel of six androgen-dependent and eight androgen-independent human prostate cancer xenografts. TRPS1 protein is expressed in all androgen-dependent xenografts, which also express AR and prostate-specific antigen (PSA). Androgen withdrawal by castration resulted in an increase in TRPS1 protein in two androgen-dependent xenografts, indicating relieved repression by action of AR. TRPS1 protein is expressed in four androgen-independent xenografts and is low or absent in the other four androgen-independent xenografts. Androgen withdrawal by castration demonstrates that TRPS1 protein levels remain the same in 1 androgen-independent xenograft, most likely due to the lack of AR expression. These data show that TRPS1 protein expression is regulated by androgens via the AR in human prostate cancer xenografts. Analysis of TRPS1 mRNA expression in normal and tumour tissue of the prostate and 18 other human tissues, showed that TRPS1 had the highest mRNA expression levels in normal and tumour tissues of breast. In addition, high TRPS1 mRNA and protein expression levels were observed in four out of five human breast cancer cell lines. In conclusion, TRPS1 protein expression is down-regulated by androgens in human prostate cancer, and analysis of TRPS1 mRNA expression levels in several human tissues showed that the highest levels were observed in normal and tumour breast tissue.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Metribolone/pharmacology , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Testosterone Congeners/pharmacology , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred Strains , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , Repressor Proteins , Tissue Distribution , Transcription Factors , Transplantation, Heterologous
19.
Clin Endocrinol (Oxf) ; 61(5): 573-81, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15521959

ABSTRACT

OBJECTIVE: Sensitivity to glucocorticoids within the normal population is highly variable and partly determined by polymorphisms in the glucocorticoid receptor (GR) gene (NR3C1). We investigated the exact sequence alteration of a TthIIII polymorphism in the GR gene, whether it is associated with glucocorticoid sensitivity, and its relationship to 3 polymorphisms of the GR gene (N363S, BclI, ER22/23EK). DESIGN: Two dexamethasone (DEX) suppression tests were performed with 1 and 0.25 mg DEX, respectively. PATIENTS: We genotyped a random subgroup of 209 participants of the Rotterdam Study, a population-based study in the elderly. MEASUREMENTS: Anthropometric parameters, cortisol, insulin and glucose levels, and lipid concentrations were measured. RESULTS: We identified the TthIIII polymorphism as a C to T mutation, 3807 bp upstream from the mRNA start site. We found 39.7% CC-carriers, 44.5% CT-carriers, and 15.8 % TT-carriers. No differences were found between TthIIII genotypes in sensitivity to DEX, baseline cortisol, insulin, glucose or cholesterol levels, or in anthropometric variables. However, all ER22/23EK-carriers also carried the TthIIII T-allele, and carriers of both these polymorphisms had a significantly smaller cortisol suppression after 1 mg DEX, lower fasting insulin levels, and lower total and low-density lipoprotein (LDL) cholesterol levels than TthIIII T carriers without the ER22/23EK variant and noncarriers. No interaction was found between the TthIIII variant and N363S or BclI polymorphisms. CONCLUSIONS: The TthIIII polymorphism is not functional by itself. However, the ER22/23EK polymorphism is without exception linked to the TthIIII T polymorphism and this haplotype is associated with a relative resistance to glucocorticoids, and a healthy metabolic profile.


Subject(s)
Polymorphism, Genetic , Promoter Regions, Genetic , Receptors, Glucocorticoid/genetics , Aged , Aged, 80 and over , Analysis of Variance , Blood Glucose/analysis , Cholesterol/blood , Cholesterol, LDL/blood , Dexamethasone/pharmacology , Female , Glucocorticoids/pharmacology , Heterozygote , Humans , Hydrocortisone/blood , Insulin/blood , Male , Middle Aged , Sequence Analysis, DNA
20.
Am J Med ; 117(3): 158-62, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15276593

ABSTRACT

PURPOSE: We recently demonstrated that a polymorphism in codons 22 and 23 of the glucocorticoid receptor gene is associated with relative glucocorticoid resistance, greater insulin sensitivity, and lower total and low-density lipoprotein cholesterol levels. In the present study, we investigated whether the ER22/23EK polymorphism is associated with survival, cholesterol levels, and two predictors of mortality: serum C-reactive protein and interleukin 6 levels. METHODS: We studied 402 men (mean [+/- SD] age, 77.8 +/- 3.6 years). C-reactive protein was measured by a highly sensitive method using a latex-enhanced immunoephelometric assay. Interleukin 6 was determined by a commercially available immulite assay. RESULTS: After a follow-up of 4 years, 73 (19%) of 381 noncarriers died, while none of the 21 ER22/23EK carriers had died (P = 0.03). C-reactive protein levels were about 50% lower in ER22/23EK carriers (P = 0.01). There were no differences in interleukin 6 levels. CONCLUSION: Carriers of the ER22/23EK polymorphism have better survival than noncarriers, as well as lower C-reactive protein levels.


Subject(s)
C-Reactive Protein/analysis , Longevity , Polymorphism, Genetic , Receptors, Glucocorticoid/genetics , Aged , Codon , Follow-Up Studies , Heterozygote , Humans , Interleukin-6/blood , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...