Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
2.
Mol Cell Endocrinol ; 414: 168-76, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26123590

ABSTRACT

Testosterone can be converted into androstenedione (4-dione) by 17ß-hydroxysteroid dehydrogenase (HSD) activity likely performed by 17ß-HSD type 2. Our objective was to evaluate the rate of testosterone conversion to 4-dione as well as expression and localization of 17ß-HSD type 2 in omental (OM) vs. subcutaneous (SC) adipose tissues of men. Formation of 4-dione from testosterone was significantly higher in homogenates (p ≤ 0.001) and explants (p ≤ 0.01) of OM than SC tissue. Microscopy analyses and biochemical assays in cell fractions localized the enzyme in the vasculature/endothelial cells of adipose tissues. Conversion of testosterone to 4-dione was weakly detected in most OM and/or SC preadipocyte cultures. Positive correlations were found between 17ß-HSD type 2 activity in whole tissue and BMI or SC adipocyte diameter. We conclude that conversion of testosterone to 4-dione detected in abdominal adipose tissue is caused by 17ß-HSD type 2 which is localized in the vasculature of the adipose compartment.


Subject(s)
Abdominal Fat/enzymology , Androstenedione/metabolism , Estradiol Dehydrogenases/metabolism , Testosterone/metabolism , Abdominal Fat/cytology , Abdominal Fat/metabolism , Body Mass Index , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Estradiol Dehydrogenases/genetics , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/metabolism , Male , Obesity/enzymology , Obesity/metabolism , Omentum/enzymology , Omentum/metabolism , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/enzymology , Subcutaneous Fat, Abdominal/metabolism
3.
J Steroid Biochem Mol Biol ; 147: 56-69, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25448733

ABSTRACT

Over the past decade, adipose tissues have been increasingly known for their endocrine properties, that is, their ability to secrete a number of adipocytokines that may exert local and/or systemic effects. In addition, adipose tissues have long been recognized as significant sites for steroid hormone transformation and action. We hereby provide an updated survey of the many steroid-converting enzymes that may be detected in human adipose tissues, their activities and potential roles. In addition to the now well-established role of aromatase and 11ß-hydroxysteroid dehydrogenase (HSD) type 1, many enzymes have been reported in adipocyte cell lines, isolated mature cells and/or preadipocytes. These include 11ß-HSD type 2, 17ß-HSDs, 3ß-HSD, 5α-reductases, sulfatases and glucuronosyltransferases. Some of these enzymes are postulated to bear relevance for adipose tissue physiology and perhaps for the pathophysiology of obesity. This elaborate set of steroid-converting enzymes in the cell types of adipose tissue deserves further scientific attention. Our work on 20α-HSD (AKR1C1), 3α-HSD type 3 (AKR1C2) and 17ß-HSD type 5 (AKR1C3) allowed us to clarify the relevance of these enzymes for some aspects of adipose tissue function. For example, down-regulation of AKR1C2 expression in preadipocytes seems to potentiate the inhibitory action of dihydrotestosterone on adipogenesis in this model. Many additional studies are warranted to assess the impact of intra-adipose steroid hormone conversions on adipose tissue functions and chronic conditions such as obesity, diabetes and cancer.


Subject(s)
Adipose Tissue/enzymology , Hydroxysteroid Dehydrogenases/metabolism , Steroids/metabolism , Adipose Tissue/metabolism , Animals , Body Fat Distribution , Humans , Obesity/enzymology , Obesity/metabolism
4.
J Steroid Biochem Mol Biol ; 137: 176-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770321

ABSTRACT

There is some confusion in the literature about steroidogenesis in endocrine glands and steroidogenesis in peripheral intracrine tissues. The objective of the present review is to bring some clarifications and better understanding about steroidogenesis in these two types of tissues. Concerns about substrate specificity, kinetic constants and place of enzymes in the pathway have been discussed. The role of 17α-hydroxylase/17-20 lyase (CYP17A1) in the production of dehydroepiandrosterone and back-door pathways of dihydrotestosterone biosynthesis is also analyzed. This article is part of a Special Issue entitled "Synthesis and biological testing of steroid derivatives as inhibitors".


Subject(s)
Steroid 17-alpha-Hydroxylase/metabolism , Steroids/biosynthesis , Humans
5.
Endocrinology ; 154(6): 2114-28, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23554453

ABSTRACT

The sulfated neurosteroids pregnenolone sulfate (Δ(5)PS) and dehydroepiandrosterone sulfate (DHEAS) are known to play a role in the control of reproductive behavior. In the frog Pelophylax ridibundus, the enzyme hydroxysteroid sulfotransferase (HST), responsible for the biosynthesis of Δ(5)PS and DHEAS, is expressed in the magnocellular nucleus and the anterior preoptic area, two hypothalamic regions that are richly innervated by GnRH1-containing fibers. This observation suggests that GnRH1 may regulate the formation of sulfated neurosteroids to control sexual activity. Double labeling of frog brain slices with HST and GnRH1 antibodies revealed that GnRH1-immunoreactive fibers are located in close vicinity of HST-positive neurons. The cDNAs encoding 3 GnRH receptors (designated riGnRHR-1, -2, and -3) were cloned from the frog brain. RT-PCR analyses revealed that riGnRHR-1 is strongly expressed in the hypothalamus and the pituitary whereas riGnRHR-2 and -3 are primarily expressed in the brain. In situ hybridization histochemistry indicated that GnRHR-1 and GnRHR-3 mRNAs are particularly abundant in preoptic area and magnocellular nucleus whereas the concentration of GnRHR-2 mRNA in these 2 nuclei is much lower. Pulse-chase experiments using tritiated Δ(5)P and DHEA as steroid precursors, and 3'-phosphoadenosine 5'-phosphosulfate as a sulfonate moiety donor, showed that GnRH1 stimulates, in a dose-dependent manner, the biosynthesis of Δ(5)PS and DHEAS in frog diencephalic explants. Because Δ(5)PS and DHEAS, like GnRH, stimulate sexual activity, our data strongly suggest that some of the behavioral effects of GnRH could be mediated via the modulation of sulfated neurosteroid production.


Subject(s)
Dehydroepiandrosterone Sulfate/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Pregnenolone/metabolism , Amino Acid Sequence , Animals , Cell Line , Diencephalon/drug effects , Diencephalon/metabolism , Gene Expression Profiling , Gonadotropin-Releasing Hormone/pharmacology , In Situ Hybridization , Male , Microscopy, Confocal , Molecular Sequence Data , Neurons/metabolism , Pituitary Gland/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ranidae , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sulfotransferases/metabolism
6.
J Matern Fetal Neonatal Med ; 26(5): 513-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23075231

ABSTRACT

OBJECTIVE: To examine maternal insulin resistance in relationship with maternal and fetal androgen levels as well as with term placenta mRNA and protein abundance of steroidogenic enzymes implicated in androgen dynamics. METHODS: The study included 20 women with gestational diabetes mellitus and 27 controls tested using a 120 min., 75 g oral glucose tolerance test. Maternal and fetal plasma concentrations of total testosterone, dihydrotestosterone (DHT) and dehydroepiandrosterone (DHEA) were measured by high-performance gas chromatography and chemical ionization mass spectrometry at 26.1 ± 3.7 weeks of pregnancy. RESULTS: Glycemic response to oral glucose over 120 min. as well as Matsuda insulin sensitivity and HOMA insulin resistance (HOMA-IR) indices were significantly associated with maternal testosterone levels (r = 0.31, r = -0.37 and r = 0.35 respectively, p ≤ 0.05 for all). Among male offspring, a positive association between maternal and fetal testosterone levels was observed (r = 0.43, p ≤ 0.05). Testosterone levels were higher in the cord blood of newborns from insulin-resistant mothers compared to newborns from insulin-sensitive mothers (0.48 ± 0.36 nmol/L vs. 0.29 ± 0.18 nmol/L p ≤ 0.05). No difference was observed in mRNA abundance or protein expression of placental steroidogenic enzymes according to the degree of maternal insulin resistance. CONCLUSION: Our results demonstrate a possible association between fetal and maternal androgen concentrations in relationship with insulin resistance.


Subject(s)
Androgens/blood , Diabetes, Gestational/blood , Fetal Blood/chemistry , Insulin Resistance/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Adult , Aromatase/genetics , Dehydroepiandrosterone/blood , Dihydrotestosterone/blood , Female , Gestational Age , Glucose Tolerance Test , Humans , Infant, Newborn , Male , Placenta/enzymology , Pregnancy , RNA, Messenger/analysis , Steroids/biosynthesis , Testosterone/blood
7.
Breast Cancer Res Treat ; 134(2): 625-47, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22678160

ABSTRACT

Novel agents for the endocrine therapy of breast cancer are needed, especially in order to take advantage of the multiple consecutive responses observed in metastatic progressing breast cancer following previous hormone therapy, thus delaying the use of cytotoxic chemotherapy with its frequent poor tolerance and serious side effects. Acolbifene (ACOL) is a novel and unique antiestrogen which represents a unique opportunity to achieve the most potent and specific blockade of estrogen action in the mammary gland and uterus while exerting estrogen-like beneficial effects in other tissues, especially the bones. To better understand the specificity of action of ACOL, we have used Affymetrix GeneChips containing 45,000 probe sets to analyze 34,000 genes to determine the specificity of this compound compared to the pure antiestrogen fulvestrant, as well as to the mixed antagonists/agonists tamoxifen and raloxifene to block the effect of estradiol (E(2)) and to induce effects of their own on the genomic profile in the mouse mammary gland. The genes modulated by E(2) were those identified in two separate experiments and validated by quantitative real-time PCR (qPCR). Three hours after the single subcutaneous injection of E(2) (0.05 µg), the simultaneous administration of ACOL, fulvestrant, tamoxifen, and raloxifene blocked by 98, 61, 43, and 92 % the number of E(2)-upregulated genes, respectively. On the other hand, 70, 10, 25, and 55 % of the genes down-regulated by E(2) were blocked by the same compounds. Of the 128 genes modulated by E(2), 49 are associated with tumorigenesis while 22 are known to be associated with breast cancer. When used alone, ACOL modulated the smallest number of genes also influenced by E(2), namely 4 %, thus possibly explaining potential utilities of this compound in breast cancer prevention and therapy.


Subject(s)
Estradiol/physiology , Estrogens/physiology , Receptors, Estrogen/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Cluster Analysis , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Female , Fulvestrant , Gene Expression Regulation , Genes , Genes, Neoplasm , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Ovariectomy , Piperidines/pharmacology , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Transcriptome
8.
Article in English | MEDLINE | ID: mdl-22654849

ABSTRACT

The enzymatic pathways leading to the synthesis of bioactive steroids in the brain are now almost completely elucidated in various groups of vertebrates and, during the last decade, the neuronal mechanisms involved in the regulation of neurosteroid production have received increasing attention. This report reviews the current knowledge concerning the effects of neurotransmitters, peptide hormones, and neuropeptides on the biosynthesis of neurosteroids. Anatomical studies have been carried out to visualize the neurotransmitter- or neuropeptide-containing fibers contacting steroid-synthesizing neurons as well as the neurotransmitter, peptide hormones, or neuropeptide receptors expressed in these neurons. Biochemical experiments have been conducted to investigate the effects of neurotransmitters, peptide hormones, or neuropeptides on neurosteroid biosynthesis, and to characterize the type of receptors involved. Thus, it has been found that glutamate, acting through kainate and/or AMPA receptors, rapidly inactivates P450arom, and that melatonin produced by the pineal gland and eye inhibits the biosynthesis of 7α-hydroxypregnenolone (7α-OH-Δ(5)P), while prolactin produced by the adenohypophysis enhances the formation of 7α-OH-Δ(5)P. It has also been demonstrated that the biosynthesis of neurosteroids is inhibited by GABA, acting through GABA(A) receptors, and neuropeptide Y, acting through Y1 receptors. In contrast, it has been shown that the octadecaneuropetide ODN, acting through central-type benzodiazepine receptors, the triakontatetraneuropeptide TTN, acting though peripheral-type benzodiazepine receptors, and vasotocin, acting through V1a-like receptors, stimulate the production of neurosteroids. Since neurosteroids are implicated in the control of various neurophysiological and behavioral processes, these data suggest that some of the neurophysiological effects exerted by neurotransmitters and neuropeptides may be mediated via the regulation of neurosteroid production.

9.
Am J Physiol Endocrinol Metab ; 302(8): E941-9, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22275760

ABSTRACT

Adipogenesis and lipid storage in human adipose tissue are inhibited by androgens such as DHT. Inactivation of DHT to 3α-diol is stimulated by glucocorticoids in human preadipocytes. We sought to characterize glucocorticoid-induced androgen inactivation in human preadipocytes and to establish its role in the antiadipogenic action of DHT. Subcutaneous and omental primary preadipocyte cultures were established from fat samples obtained in subjects undergoing abdominal surgeries. Inactivation of DHT to 3α/ß-diol for 24 h was measured in dexamethasone- or vehicle-treated cells. Specific downregulation of aldo-keto reductase 1C (AKR1C) enzymes in human preadipocytes was achieved using RNA interference. In whole adipose tissue sample, cortisol production was positively correlated with androgen inactivation in both subcutaneous and omental adipose tissue (P < 0.05). Maximal dexamethasone (1 µM) stimulation of DHT inactivation was higher in omental compared with subcutaneous fat from men as well as subcutaneous and omental fat from women (P < 0.05). A significant positive correlation was observed between BMI and maximal dexamethasone-induced DHT inactivation rates in subcutaneous and omental adipose tissue of men and women (r = 0.24, n = 26, P < 0.01). siRNA-induced downregulation of AKR1C2, but not AKR1C1 or AKR1C3, significantly reduced basal and glucocorticoid-induced androgen inactivation rates (P < 0.05). The inhibitory action of DHT on preadipocyte differentiation was potentiated following AKR1C2 but not AKR1C1 or AKR1C3 downregulation. Specifically, lipid accumulation, G3PDH activity, and FABP4 mRNA expression in differentiated preadipocytes exposed to DHT were reduced further upon AKR1C2 siRNA transfection. We conclude that glucocorticoid-induced androgen inactivation is mediated by AKR1C2 and is particularly effective in omental preadipocytes of obese men. The interplay between glucocorticoids and AKR1C2-dependent androgen inactivation may locally modulate adipogenesis and lipid accumulation in a depot-specific manner.


Subject(s)
Adipocytes, White/drug effects , Adipogenesis/drug effects , Androgens/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hydroxysteroid Dehydrogenases/metabolism , Obesity/metabolism , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adult , Body Mass Index , Cells, Cultured , Dihydrotestosterone/metabolism , Female , Humans , Hydrocortisone/metabolism , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxysteroid Dehydrogenases/chemistry , Hydroxysteroid Dehydrogenases/genetics , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Male , Middle Aged , Molecular Targeted Therapy , Obesity/drug therapy , Obesity/pathology , RNA Interference , RNA, Small Interfering , Sex Characteristics , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology
10.
Pathol Oncol Res ; 17(3): 627-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21267686

ABSTRACT

Intratumoral biosynthesis of hormone steroids is thought to play a role in the pathogenesis and development of human breast cancer. There is evidence that glucocorticoids may inhibit the development and progression of breast cancer. 11ß-hydroxysteroid dehydrogenase (11ß-HSD) type 1 is the enzyme which converts inactive cortisone to active cortisol. In order to study the expression of 11ß-HSD type 1 in breast cancer and non-cancerous breast tissue, we have developed specific antibodies to 11ß-HSD type 1 and proceeded to localization of the enzyme in 84 specimens of breast carcinoma and adjacent non-malignant tissues by immnohistochemistry. The results were correlated with the expression of androgen receptor, estrogen receptor, progesterone receptor, glucocorticoid receptor and CDC47, a cell division marker, as well as the tumor stage, tumor size, nodal status and menopausal status. The expression of 11ß-HSD type 1 in 64% of breast cancer specimens appeared significantly lower than that observed in normal adjacent tissues (97% of cases being positive). There was no significant correlation between 11ß-HSD type 1 expression and the clinicopathological parameters studied. The decrease in 11ß-HSD type 1 expression in breast cancer as compared to that observed in the adjacent normal tissues may play a role in the development and/or progression of the cancer by modifying the intratumoral levels of glucocorticoids.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast/metabolism , Adult , Aged , Breast/pathology , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Immunoenzyme Techniques , Middle Aged , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/metabolism , Prognosis , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism
11.
Horm Mol Biol Clin Investig ; 5(3): 161-5, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-25961251

ABSTRACT

Traditional literature and textbooks generally describe that estradiol (E2) and dihydrotestosterone (DHT) are synthesized from the aromatization and 5α-reduction of testosterone (T), respectively, following a pathway in which T is an essential intermediate (Tpath). This pathway implies that the steps of aromatization and 5α-reduction follow the reaction of the androgenic 17ß-hydroxysteroid dehydrogenase (17ß-HSD) that catalyzes the conversion of 4-androstenedione (4-dione) into T, and that estrogenic 17ß-HSDs are not required. Contrary to this belief, the cloning of many estrogen-specific 17ß-HSDs and the observation of higher affinity of aromatase and 5α-reductase for 4-dione than T are strongly in favor of biosynthetic pathways in which the steps catalyzed by aromatase and 5α-reductase precede that catalyzed by 17ß-HSDs. Such pathways do not require T as an intermediate, as demonstrated by experiments using [14C]-labeled DHEA and 4-dione as substrates and incubation with SZ95 sebaceous gland, DU-145 prostate cancer and JEG-3 choriocarcinoma cell lines cultured in the presence of inhibitors of 5α-reductase and aromatase. A review of early literature about patients with testicular 17ß-HSD deficiency and of steroid metabolism appears to confirm the physiological functionality of the E2 and DHT biosynthetic pathway not requiring T as intermediate (noTpath).

12.
Article in English | MEDLINE | ID: mdl-22649387

ABSTRACT

Amphibians have been widely used to investigate the synthesis of biologically active steroids in the brain and the regulation of neurosteroid production by neurotransmitters and neuropeptides. The aim of the present review is to summarize the current knowledge regarding the neuroanatomical distribution and biochemical activity of steroidogenic enzymes in the brain of anurans and urodeles. The data accumulated over the past two decades demonstrate that discrete populations of neurons and/or glial cells in the frog and newt brains express the major steroidogenic enzymes and are able to synthesize de novo a number of neurosteroids from cholesterol/pregnenolone. Since neurosteroidogenesis has been conserved during evolution from amphibians to mammals, it appears that neurosteroids must play important physiological functions in the central nervous system of vertebrates.

13.
Prog Brain Res ; 181: 177-92, 2010.
Article in English | MEDLINE | ID: mdl-20478438

ABSTRACT

There is an increasing number of differences reported between the steroidogenesis pathways described in the traditional literature related to gonadal steroidogenesis and the more recent observations achieved using new technologies, especially molecular cloning, pangenomic expression studies, precise quantification of mRNA expression using real-time PCR, use of steroidogenic enzymes stably transfected in cells, detailed enzymatic activity analysis in cultured cell lines and mass spectrometry analysis of steroids. The objective of this chapter is to present steroidogenesis in the light of new findings that demonstrate pathways of biosynthesis of estradiol (E(2)) and dihydrotestosterone (DHT) from adrenal dehydroepiandrosterone (DHEA) in peripheral intracrine tissues which do not involve testosterone as intermediate as classically found in the testis and ovary. Steroidogenic enzymes different from those of the ovary and testis act in a tissue-specific manner to catalyze the transformation of DHEA into active sex steroids. These new pathways are especially important in post-menopausal women where all estrogens and practically all androgens are made at their site of action in peripheral tissues from DHEA, the precursor of adrenal origin. In men, on the other hand, from 40 to 50% of androgens are made in peripheral tissues from adrenal DHEA, thus indicating the major importance of the intracrine pathways in both men and women. We also examine the molecular evolution of steroidogenic enzymes which explains the major differences in steroid metabolism observed between laboratory animals and humans.


Subject(s)
Dihydrotestosterone/metabolism , Estradiol/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Adrenal Glands/chemistry , Adrenal Glands/metabolism , Animals , Dehydroepiandrosterone/metabolism , Estradiol/genetics , Evolution, Molecular , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Male , Ovary/chemistry , Ovary/metabolism , Steroid Hydroxylases/classification , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Testis/chemistry , Testis/metabolism
14.
J Steroid Biochem Mol Biol ; 122(1-3): 28-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20206259

ABSTRACT

Adipose tissue glucocorticoid action relies on local enzymatic interconversion and glucocorticoid receptor (GR) availability. 11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1), 2 (11ß-HSD2) and hexose-6-phosphate dehydrogenase (H6PDH) are likely involved in glucocorticoid activation/inactivation within adipose tissue. We examined adipose tissue mRNA expression of genes related to glucocorticoid action and their association with total and visceral adiposity. Messenger RNA was measured in paired subcutaneous and omental fat samples obtained from 56 women (age: 47.3 ± 4.8 years, BMI: 27.1 ± 5.2 kg/m(2)) undergoing gynaecological surgery. Expression levels of 11ß-HSD2, H6PDH and GRα were higher in omental adipose tissue while 11ß-HSD1 expression was similar between fat compartments. Subcutaneous and omental 11ß-HSD1 mRNA abundances were positively associated with total and visceral adiposity whereas omental H6PDH mRNA abundance was negatively associated with these measures. Only omental 11ß-HSD1 mRNA expression remained significantly associated with visceral adipose tissue area following statistical adjustment for fat mass, age and menopausal status. Omental 11ß-HSD1 mRNA expression explained 19.1% of the variance in visceral adipose tissue area. Omental fat tissue 11ß-HSD-1 protein and cortisol levels were higher in visceral obese women, supporting findings obtained with 11ß-HSD-1 mRNA. These results suggest that among the transcripts examined only omental 11ß-HSD1 is independently associated with visceral obesity in women.


Subject(s)
Gene Expression Regulation, Enzymologic , Glucocorticoids/metabolism , Intra-Abdominal Fat/physiology , Subcutaneous Fat/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adult , Blotting, Western , Carbohydrate Dehydrogenases/biosynthesis , Carbohydrate Dehydrogenases/genetics , Female , Humans , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/metabolism , Linear Models , Middle Aged , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Subcutaneous Fat/enzymology , Subcutaneous Fat/metabolism
15.
J Chem Neuroanat ; 39(1): 35-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19665548

ABSTRACT

The occurrence of several enzymes responsible for the biosynthesis of neurosteroids in the brain of adult frogs is now firmly established but the expression of these enzymes during ontogenesis has not yet been investigated. In the present report, we describe the immunohistochemical distribution and biological activity of 3beta-hydroxysteroid dehydrogenase (3beta-HSD) and 5alpha-reductase (5alpha-R) in the brain of the European green frog, Rana esculenta, during larval development. The spatio-temporal distribution of 3beta-HSD and 5alpha-R immunoreactivities in the tadpole brain was generally different, although these two enzymes were occasionally detected in the same areas such as the olfactory bulbs and cerebellum. Identification of neurons based on their morphological aspect as well as labeling of astrocytes with an antiserum against glial fibrillary acidic protein (GFAP) revealed that, in the tadpole brain, 3beta-HSD- and 5alpha-R-immunoreactive materials were contained in both neurons and glial cells. Incubation of tadpole brain explants with [(3)H]-pregnenolone resulted in the formation of several tritiated steroids including progesterone, 17-hydroxyprogesterone, androstenedione, 5alpha-dihydroprogesterone and 5alpha-dihydrotestosterone. The present study provides the first immunocytochemical mapping of two key steroidogenic enzymes in the developing frog brain. The data also indicate that neurosteroid biosynthesis occurs in the brain of tadpoles, as previously shown for adult amphibians, birds and mammals. The transient expression of steroidogenic enzymes in several regions of the tadpole brain suggests that, in amphibians, neurosteroids may be implicated in neurotrophic activities during larval development.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Brain/enzymology , Rana esculenta/metabolism , Animals , Immunohistochemistry , Larva
17.
Horm Mol Biol Clin Investig ; 2(3): 293-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-25961201

ABSTRACT

5α-Reductases are crucial enzymes involved in the biosynthesis of dihydrotestosterone, the most potent natural androgen. To date, three types of 5α-reductases, chronologically named types 1, 2 and 3 5α-reductases (SRD5a-1, 2 and 3) have been described. In the present paper, we characterized the activity and compared the mRNA expression levels of SRD5a-3 with those of SRD5a-1 and 2 in various human tissues, and determined its sensitivity to finasteride and dutasteride. We have established HEK-293 cell line that stably expressed SRD5a-3 for studying its activity and the inhibitory effect of finasteride, using [14C]labeled steroids. mRNA expression levels were quantified using real-time PCR in many male and female human tissues including the prostate, adipose tissue, mammary gland, as well as breast and prostate cancer cell lines. Incubation of HEK-SRD5a-3 cells with [14C]4-androstenedione and [14C]testosterone allowed us to show that SRD5a-3 can catalyze very efficiently both substrates 4-androstenedione and testosterone into 5α-androstanedione and dihydrotestosterone, respectively. We observed that the affinity of the enzyme for 4-androstenedione is higher than for testosterone. The activity of SRD5a-3 and SRD5a-2 are similarly sensitive to finasteride, whereas dutasteride is a much more potent inhibitor of SRD5a-3 than SRD5a-2. Tissue distribution analysis shows that SRD5a-3 mRNA expression levels are higher than those of SRD5a-1 and SRD5a-2 in 20 analyzed tissues. In particular, it is highly expressed in the skin, brain, mammary gland and breast cancer cell lines, thus suggesting that SRD5a-3 could play an important role in the production of androgens in these and other peripheral tissues.

18.
Horm Mol Biol Clin Investig ; 2(3): 311-8, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-25961203

ABSTRACT

17ß-Hydroxysteroid dehydrogenases (17ß-HSDs) are enzymes issued from convergent evolution of activity from various ancestral genes having different functions. Type 12 17ß-HSD (17ß-HSD12) was described as a bifunctional enzyme, involved in the biosynthesis of estradiol (E2) and the elongation of very long chain fatty acid (VLCFA). It catalyzes selectively the transformation of estrone (E1) into estradiol (E2) in human and primates, whereas in the mouse and Caenorhabditis elegans the enzyme catalyzes the 17ß-reduction of both androgens and estrogens. It is also able to catalyze the reduction of 3-keto-acylCoA into 3-hydroxy-acylCoA in the elongation cycle of VLCFA biosynthesis. To further understand the physiological role of 17ß-HSD12, we performed targeted disruption of the Hsd17b12 gene by substituting exons 8 and 9 that contain the active site with a neomycin cassette. The data indicate that heterozygous (HSD17B12+/-) mice are viable with reduced levels of sex steroids, whereas homozygous (HSD17B12-/-) mice show embryonic lethality. The present data are in agreement with the bifunctional activities of 17ß-HSD12 suggesting that the VLCFA elongation activity, having its origin in the yeast, is most probably responsible for embryonic lethality in HSD17B12-/-, whereas the more recently acquired 17ß-HSD12 activity is responsible for reduced sex steroid levels in HSD17B12+/-.

19.
Horm Mol Biol Clin Investig ; 3(2): 411-5, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-25961212

ABSTRACT

Intratumoral biosynthesis of sex steroids is thought to play a role in the pathogenesis and development of human breast cancer. There is evidence that androgens can inhibit the development and progression of breast cancer. Among the enzymes involved in the biosynthesis of androgens, 5α-reductase plays a key role by reducing testosterone to dihydrotestosterone, the most potent androgen. Two isoforms of 5α-reductase have been characterized and 5α-reductase type 1 is predominant in breast cancer tissue. We developed specific antibodies to 5α-reductase type 1 and studied the expression of the enzyme in 84 specimens of breast carcinoma and adjacent non-malignant tissues by immunohistochemistry. The results were correlated with the expression of androgen receptor, estrogen receptor α, progesterone receptor and CDC47, a cell division marker as well as the tumor stage, tumor size, nodal status and menopausal status. The expression of 5α-reductase type 1 in 61% of breast cancer specimens appeared significantly lower than that observed in normal adjacent tissues (87% of cases being positive). There was no significant correlation between 5α-reductase type 1 expression and the clinicopathological parameters studied. The decrease in 5α-reductase type 1 expression in breast cancer as compared to that observed in the adjacent normal tissues could play a role in the development and/or progression of the cancer by modifying the intratumoral levels of androgens.

20.
Horm Mol Biol Clin Investig ; 1(2): 67-72, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-25961972

ABSTRACT

BACKGROUND: Although it is well recognized that 5α-reductases possess higher affinity for 4-androstenedione than testosterone, and the affinity of 4-androstenedione is higher for 5α-reductases than 17ß-hydroxysteroid dehydrogenases, it is generally believed that dihydrotestosterone is necessarily produced by the transformation of testosterone into dihydrotestosterone, suggesting that the step catalyzed by 17ß-hydroxysteroid dehydrogenase precedes the step catalyzed by 5α-reductase. This interpretation is in contradiction with the enzymatic kinetic law that suggests that the 5α-reduction step that catalyzes the transformation of 4-dione into 5α-androstane-3,17-dione precedes the 17keto-reduction step. MATERIALS AND METHODS: To verify which of these two pathways is operative, we quantified mRNA expression levels of steroidogenic enzymes in prostate carcinoma DU-145 cells by real-time PCR and determined the metabolites produced after incubation with [14C]4-dione in the presence and absence of a 5α-reductase inhibitor and analyzed the metabolites produced by thin layer chromatography and HPLC. RESULTS: Real-time PCR analysis strongly suggests that the new type 3 5α-reductase is responsible for 5α-reductase activity in DU-145 cells. Steroid profile analysis shows that in the absence of inhibitor 5α-androstanedione is first produced, followed by the production of androsterone and dihydrotestosterone. The concentration of testosterone was not detectable. In the presence of Finasteride, an inhibitor of 5α-reductase, there was no transformation of 4-androstenedione and also there was no production of testosterone. The present data clearly indicate that the biosynthesis of dihydrotestosterone in DU-145 cells does not require testosterone as intermediate, and the step catalyzed by 5α-reductase precedes the step catalyzed by 17ß-hydroxysteroid dehydrogenase.

SELECTION OF CITATIONS
SEARCH DETAIL
...