Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Horm Mol Biol Clin Investig ; 10(3): 293-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-25436687

ABSTRACT

Abstract Interest has developed into 7-hydroxylated derivatives of 3ß-hydroxylated C19-steroids, such as dehydroepiandrosterone (DHEA) and epiandrosterone because of their effects on inflammation, immune response, and cell repair. These steroids are not currently available from commercial sources, and it is necessary to produce them for relevant studies. We report here the chemical and biochemical approaches that were used for their production. Simplified chemical approaches lead to the production of 7α-/7ß-hydroxy-DHEA and 7ß-hydroxy-epiandrosterone in gram quantities, which are made available to researchers. Biochemical approaches were used to produce isotope-labeled compounds. Thus, 2H-, 3H-, and 14C-labeled 7α-/7ß-hydroxy-DHEA and 7ß-hydroxy-epiandrosterone could be produced in quantities sufficient for use in investigations into their mode of action.

2.
Horm Mol Biol Clin Investig ; 9(3): 179-89, 2012 Apr.
Article in English | MEDLINE | ID: mdl-25436707

ABSTRACT

Abstract Successive action of cytochrome P450-7B1 (CYP7B1) and 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) on 3ß-hydroxysteroids such as DHEA and epiandrosterone leads to the production of cytoprotective 7ß-hydroxylated derivatives. Investigation of the presence of these enzymes in human tissues could be carried out on commercially available human tissue arrays with use of antibodies specific to CYP7B1 and 11ß-HSD1 for immunohistochemistry. Both enzymes were detected mainly in tissues of endodermic and ectodermic origin which are prone to undergo inflammation. As low doses of the 7ß-hydroxylated derivatives of DHEA and epiandrosterone trigger the resolution of inflammation and tissue repair, CYP7B1 and 11ß-HSD1 tissue contents may reflect the tissue ability for reparation after pathological conditions.

3.
Horm Mol Biol Clin Investig ; 7(1): 295-301, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-25961267

ABSTRACT

In order to develop an immunoassay of 7ß-hydroxy-epiandrosterone, a stereoselective synthesis of a specific hapten, 7ß-hydroxy-19-oxo-androstan 19-(O-carboxymethyl)oxime (17), was performed. This synthesis was achieved in 16% overall yield starting from the well-known 3ß-acetoxy-19-hydroxy-5-androsten-17-one (1). After coupling of the alkyl oxime moiety, an allylic oxidation of the C-7 carbon under mild conditions followed by two selective reductions established all the functionalities of the final compound 17.

4.
Steroids ; 76(1-2): 28-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20727907

ABSTRACT

The synthesis of 7ß-hydroxy-epiandrosterone (6) possessing strong anti-inflammatory properties was achieved starting from 3ß-acetoxy-17,17-(ethylenedioxy)-5-androsten (1). This approach involved as a main step an allylic oxidation of the C-7 followed by two reduction reactions of the double bond and of the carbonyl group. This stereoselective synthesis in 5 steps gave 7ß-hydroxy-epiandrosterone in 63% overall yield.


Subject(s)
Androsterone/analogs & derivatives , Androsterone/chemical synthesis , Androsterone/chemistry , Molecular Conformation , Stereoisomerism
5.
Horm Mol Biol Clin Investig ; 4(1): 489-98, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-25961225

ABSTRACT

Numerous studies investigated the effects of pharmacological doses of DHEA in animals. Among protective effects, antiglucocorticoid potencies, triggering and modulation of immunity and anticancerous effects were reported. Because DHEA levels decrease in aging humans, this steroid has been assayed as replacement therapy in elderly volunteers without striking evidence for beneficial effects. Examination of the investigations carried out in animals lead to suspect that, rather than DHEA, its metabolites produced in tissues could be responsible for some of the observed effects. Known as the "mother steroid", DHEA is a precursor for androgenic and estrogenic steroid hormones. In addition, DHEA is hydroxylated at the 7α position by the cytochrome P450 7B1 (CYP7B1), and the 7α-hydroxy-DHEA produced is a substrate for the 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) which converts it into 7ß-hydroxy-DHEA. Both 7-hydroxylated metabolites were shown to favor the onset of immunity in mice and the activation of memory T cells in humans. Other DHEA and testosterone-derived metabolites, namely epiandrosterone and 5α-androstane-3ß,17ß-diol, are also substrates for the CYP7B1 and their 7α-hydroxylated products were also converted into the 7ß epimer by the 11ß-HSD1. When assayed at doses 104 lower than DHEA, 7ß-hydroxy-epiandrosterone was shown to shift the prostaglandin metabolism patterns from prostaglandin E2 (PGE2) to PGD2 production, thus triggering the resolution of inflammation. In addition, 7ß-hydroxy-epiandrosterone (1 nM) exerted the same effects as tamoxifen (1 µM) on the proliferation of MCF-7 and MDA-231 human breast cancer cells. These findings suggest that the observed effects of 7ß-hydroxy-epiandrosterone could be mediated by estrogen receptors. This overview of recent research implies that DHEA does not act directly and that its effects are due to its metabolites when produced in tissues. Treatments with DHEA should take into account the target tissue abilities to produce the desired metabolites through the two key enzymes, CYP7B1 and 11ß-HSD1.

6.
Horm Mol Biol Clin Investig ; 1(1): 11-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-25961967

ABSTRACT

Inflamed tissues produce both prostaglandins (PGs) and 7α-hydroxylated derivatives of native circulating 3ß-hydroxysteroids. These 7α-hydroxysteroids are in turn transformed into 7ß-hydroxylated epimers by 11ß-hydroxysteroid dehydrogenase type 1 in the tissue. 7ß-Hydroxy-epiandrosterone (7ß-hydroxy-EpiA) affects PG production in two models of inflammation, dextran sodium sulfate-induced colitis in the rat and TNF-α-induced activation of PG production and PG synthase expression in cultured human peripheral blood monocytes (hPBMC). Treatment with 7ß-hydroxy-EpiA led to a shift from high to low colonic PGE2 levels and from low to high 15-deoxy-Δ12-14-PGJ2 (15d-PGJ2) levels, together with changes in the expression of the respective PG synthases and resolution of colonic inflammation. Addition of 7ß-hydroxy-EpiA to hPBMC also changed the expression of PG synthases and decreased PGE2 while increasing 15d-PGJ2 production. These effects were only observed with 7ß-hydroxy-EpiA and not with 7α-hydroxy- or 7ß-hydroxy-dehydroepiandrosterone (7α-hydroxy-DHEA and 7ß-hydroxy-DHEA). 15d-PGJ2, which is the native ligand for peroxisome proliferator-activated receptor subtype γ, contributes to cell protection and to the resolution of inflammation. Our results therefore suggest that 7ß-hydroxy-EpiA may facilitate inflammatory resolution by shifting PG production from PGE2 to PGD2 and 15d-PGJ2. The finding that 7ß-hydroxy-EpiA was effective at nM concentrations, whereas the two structurally closely related hydroxysteroids 7α-hydroxy-DHEA and 7ß-hydroxy-DHEA were inactive suggests that the effects of 7ß-hydroxy-EpiA are specific to this steroid and may be mediated by a specific receptor.

7.
Neuro Endocrinol Lett ; 30(3): 368-72, 2009.
Article in English | MEDLINE | ID: mdl-19855361

ABSTRACT

OBJECTIVE: Dehydroepiandrosterone is a long established neuroactive steroid. Some authors documented that 7-oxygenated derivatives of this steroid may be responsive at least by part for its physiological activity. METHODS: In the ventricular cerebrospinal fluid obtained from 15 patients with hydrocephalus (8 postmenopausal women and 7 men) potentially neuroactive steroid 7-oxygenated derivatives of dehydroepiandrosterone were quantified using gas chromatography/mass spectrometry. RESULTS: Besides free dehydroepiandrosterone 7-oxygenated steroids such as 7alpha- and 7beta-hydroxy-dehydroepiandrosterone, 5-androstene-3beta,7alpha,17beta-triol and 5-androstene-3beta,7beta,17beta-triol in picomolar concentration in serum and cerebrospinal fluid were found. CONCLUSION: Dehydroepiandrosterone and its 7-oxygenated derivatives are present in ventricular cerebrospinal fluid in concentration 2 -100 times lower than in serum.


Subject(s)
Androstenols/cerebrospinal fluid , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/cerebrospinal fluid , Hydrocephalus/cerebrospinal fluid , Adult , Androstenols/blood , Dehydroepiandrosterone/blood , Female , Humans , Hydrocephalus/blood , Hydrocephalus/surgery , Male , Mass Spectrometry
8.
J Steroid Biochem Mol Biol ; 114(1-2): 57-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19167490

ABSTRACT

Cytochrome P4507B1 7alpha-hydroxylates dehydroepiandrosterone (DHEA), epiandrosterone (EpiA) and 5alpha-androstane-3beta,17beta-diol (Adiol). 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) interconverts 7alpha- and 7beta-forms. Whether the interconversion proceeds through oxido-reductive steps or epimerase activity was investigated. Experiments using [(3)H]-labelled 7beta-hydroxy-DHEA, 7beta-hydroxy-EpiA and 7beta-hydroxy-Adiol showed the (3)H-label to accumulate in the 7-oxo-DHEA trap but not in 7-oxo-EpiA or 7-oxo-Adiol traps. Computed models of 7-oxygenated steroids docked in the active site of 11beta-HSD1 either in a flipped or turned form relative to cortisone and cortisol. 7-Oxo-steroid reduction in 7alpha- or 7beta-hydroxylated derivatives resulted from either turned or flipped forms. 11beta-HSD1 incubation in H(2)(18)O medium with each 7-hydroxysteroid did not incorporate (18)O in 7-hydroxylated derivatives of EpiA and Adiol independently of the cofactor used. Thus oxido-reductive steps apply for the interconversion of 7alpha- and 7beta-hydroxy-DHEA through 7-oxo-DHEA. Epimerization may proceed on the 7-hydroxylated derivatives of EpiA and Adiol through a mechanism involving the cofactor and Ser(170). The physiopathological importance of this epimerization process is related to 7beta-hydroxy-EpiA production and its effects in triggering the resolution of inflammation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Steroid Hydroxylases/metabolism , Steroids , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Androstane-3,17-diol/chemistry , Androstane-3,17-diol/metabolism , Androsterone/chemistry , Androsterone/metabolism , Catalytic Domain , Cytochrome P450 Family 7 , Dehydroepiandrosterone/chemistry , Dehydroepiandrosterone/metabolism , Humans , Hydroxylation , Molecular Structure , Oxidation-Reduction , Steroid Hydroxylases/genetics , Steroids/chemistry , Steroids/metabolism
9.
Steroids ; 73(11): 1148-59, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18555503

ABSTRACT

7alpha-Hydroxy-DHEA, 7beta-hydroxy-DHEA and 7beta-hydroxy-EpiA are native metabolites of dehydroepiandrosterone (DHEA) and epiandrosterone (EpiA). Since numerous steroids are reported to interfere with inflammatory and immune processes, our objective was to test the effects of these hydroxysteroids on prostaglandin (PG) production and related enzyme gene expression. Human peripheral blood monocytes were cultured for 4 and 24 h in the presence of each of the steroids (1-100 nM), with and without addition of TNF-alpha (10 ng/mL). Levels of PGE(2), PGD(2) and 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) were measured in the incubation medium, and cell content of cyclooxygenase (COX-2), and PGE and PGD synthases (m-PGES1, H-PGDS, L-PGDS), and peroxisome proliferator activated receptor (PPAR-gamma) was assessed by quantitative RT-PCR and Western blots. Addition of TNF-alpha resulted in elevated PG production and increased COX-2 and m-PGES1 levels. Among the three steroids tested, only 7beta-hydroxy-EpiA decreased COX-2, m-PGES1 and PPAR-gamma expression while markedly decreasing PGE(2) and increasing 15d-PGJ(2) production. These results suggest that 7beta-hydroxy-EpiA is a native trigger of cellular protection through simultaneous activation of 15d-PGJ(2) and depression of PGE(2) synthesis, and that these effects may be mediated by activation of a putative receptor, specific for 7beta-hydroxy-EpiA.


Subject(s)
Androsterone/analogs & derivatives , Dinoprostone/biosynthesis , Gene Expression/drug effects , Monocytes/metabolism , Prostaglandin D2/biosynthesis , Androsterone/chemistry , Androsterone/metabolism , Androsterone/pharmacology , Cells, Cultured , Culture Media/analysis , Culture Media/chemistry , Cyclooxygenase 2/analysis , Cyclooxygenase 2/biosynthesis , Dinoprostone/analysis , Dinoprostone/chemistry , Dinoprostone/genetics , Dose-Response Relationship, Drug , Humans , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/biosynthesis , Lipocalins/analysis , Lipocalins/biosynthesis , Models, Biological , Molecular Structure , Monocytes/drug effects , PPAR gamma/analysis , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/analysis , Prostaglandin D2/chemistry , Prostaglandin D2/genetics , Prostaglandin-E Synthases , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
10.
J Steroid Biochem Mol Biol ; 110(3-5): 255-62, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18502118

ABSTRACT

High dose levels of dehydroepiandrosterone and its 7-hydroxylated derivatives have been shown to reduce oxidative stress and inflammatory responses in dextran sodium sulfate (DSS)-induced colitis in rats. Another endogenous steroid, 7beta-hydroxy-epiandrosterone (7beta-hydroxy-EpiA) has been shown to exert neuroprotective effects at much smaller doses. Our aims were to evaluate whether 7beta-hydroxy-EpiA pre-treatment prevents DSS-induced colitis and to determine whether the effects involve changes in anti-inflammatory prostaglandin (PG) D(2) and 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) levels. Rats were administered 0.01, 0.1 and 1mg/kg 7beta-hydroxy-EpiA i.p. once a day for 7 days. Thereafter, colitis was induced by administration of 5% DSS in drinking water for 7 days. Levels of the PGs and the expression of cyclooxygenase (COX-2) and PG synthases were assessed during the course of the experiment. Administration of 7beta-hydroxy-EpiA caused a transient increase in COX-2 and PGE synthase expression within 6-15h and augmented colonic tissue levels of 15d-PGJ(2) levels starting at day 2. Treatment with DSS resulted in shortened colon length, depleted mucus in goblet cells and induced oxidative stress. COX-2 and mPGES-1 synthase expression were enhanced and accompanied by increased PGE(2), D(2) and 15d-PGJ(2) production. Although all dose levels of 7beta-hydroxy-EpiA reduced PGE(2) production, only the lowest dose (0.01mg/kg) of the steroid completely prevented colitis damage and tissue inflammation. 7beta-Hydroxy-EpiA pre-treatment prevents the occurrence of DSS-induced colitis through a shift from PGE(2) to PGD(2) production, associated with an early but transient increase in COX-2 expression and a sustained increase in the production of the anti-inflammatory prostaglandin 15d-PGJ(2).


Subject(s)
Androsterone/analogs & derivatives , Androsterone/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Prostaglandins/metabolism , Algorithms , Androsterone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/metabolism , Colitis/prevention & control , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytoprotection/drug effects , Dextran Sulfate , Drug Evaluation, Preclinical , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Male , Prostaglandins/blood , Rats , Rats, Wistar
11.
Steroids ; 72(13): 855-64, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17825335

ABSTRACT

Several studies have shown that the native 7alpha-hydroxy-dehydroepiandrosterone (7alpha-hydroxy-DHEA) is a substrate for the human 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) which converts the 7alpha- into the 7beta-epimer through an oxido-reduction process. Research on the 11beta-HSD1 has investigated its function and structure through using native glucocorticoid substrates and known inhibitors. Other steroid substrates are also of interest. Among testosterone metabolites, 5alpha-androstane-3beta,17beta-diol (Adiol) is a substrate for the cytochrome P450 7B1 which produces 5alpha-androstane-3beta,7alpha,17beta-triol (7alpha-Adiol). This steroid may be a substrate for the 11beta-HSD1. We used recombinant yeast-expressed 11beta-HSD1 with NADP(H)-regenerating systems for examining the products obtained after incubation with 7alpha-Adiol, 7beta-Adiol or 7-oxo-Adiol. Oxidative conditions for the 11beta-HSD1 provided no trace of 7-oxo-Adiol but the inter-conversion of 7alpha- and 7beta-hydroxy-Adiol with V(max)/K(M) (pmol min(-1) microg(-1)/microM) values of 2 and 0.5, respectively. This state was maintained under reductive conditions. The use of a 7-oxo-Adiol substrate under reductive conditions led to the production of both 7alpha- and 7beta-hydroxy-Adiol with V(max)/K(M) values of 3.43 and 0.22, respectively. These findings support the hypothesis that the oxido-reductase and epimerase activities of 11beta-HSD1 depend on the positioning of the steroid substrates within the active site and may provide insight into its fine structure and mechanism of action.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Androstane-3,17-diol/analogs & derivatives , Androstane-3,17-diol/chemistry , Humans , Kinetics , Substrate Specificity , Water/chemistry
12.
J Steroid Biochem Mol Biol ; 105(1-5): 159-65, 2007.
Article in English | MEDLINE | ID: mdl-17624766

ABSTRACT

The human 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes both the NADP(H)-dependent oxido-reduction of cortisol and cortisone and the inter-conversion of 7alpha- and 7beta-hydroxy-dehydroepiandrosterone (DHEA) through a 7-oxo-DHEA intermediate. As shown with human liver and intestine fractions, 7alpha-hydroxy-epiandrosterone (7alpha-hydroxy-EpiA) and 7beta-hydroxy-EpiA were readily inter-converted with no evidence for a 7-oxo-EpiA intermediate. Whether this inter-conversion resulted from action of the 11beta-HSD1 or from an unknown epimerase is unresolved. Furthermore, whether these steroids could inhibit the cortisol-cortisone oxido-reduction remains a question. The recombinant human 11beta-HSD1 was used to test these questions. NADP(+) supplementation only provided the production of 7beta-hydroxy-EpiA out of 7alpha-hydroxy-EpiA with a V(max)/K(M) ratio at 0.1. With NADPH supplementation, both 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA were formed in low amounts from 7beta-hydroxy-EpiA and 7alpha-hydroxy-EpiA, respectively. These inter-conversions occurred without a trace of the putative 7-oxo-EpiA intermediate. In contrast, the 7-oxo-EpiA substrate was efficiently reduced into 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA, with V(max)/K(M) ratios of 23.6 and 5.8, respectively. Competitive and mixed type inhibitions of the 11beta-HSD1-mediated cortisol oxidation were exerted by 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA, respectively. The 11beta-HSD1-mediated cortisone reduction was inhibited in a competitive manner by 7-oxo-EpiA. These findings suggest that the active site of the human 11beta-HSD1 may carry out directly the epimeric transformation of 7-hydroxylated EpiA substrates. The low amounts of these steroids in human do not support a physiological importance for modulation of the glucocorticoid status in tissues.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Androsterone/metabolism , Enzyme Inhibitors/pharmacology , Base Sequence , Cortisone/metabolism , DNA Primers , Humans , Hydrocortisone/metabolism , Kinetics , Substrate Specificity
13.
Prostate ; 67(13): 1439-46, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17639508

ABSTRACT

BACKGROUND: Cytochrome P450 (CYP) 7B1 is involved in many metabolic processes including androgen metabolism. Cytochrome P450 (CYP) 7B1 is expressed within the prostate and may determine the levels of the natural estrogen receptor beta (ERbeta) ligand 5alpha-androstane-3beta,17beta-diol (3betaAdiol) available and hence affect the regulation of prostate proliferation. We hypothesized that CYP7B1 expression is increased in prostate tumors and that promoter methylation contributes to the regulation of CYP7B1 expression in human prostate tissue. METHODS: Expression of the CYP7B1 gene and protein in clinical prostate tissues and prostate cancer cell lines were investigated using real-time PCR and immunohistochemistry. The methylation status of the CYP7B1 gene was analyzed using methylation-specific PCR (MSP). RESULTS: The immunohistochemical results demonstrate that high expression of CYP7B1 protein occurs in high-grade prostatic intraepithelial neoplasia (PIN) and adenocarcinomas. The ERbeta/CYP7B1 mRNA ratio was significantly lower in tumor compared to the non-tumor area. The MSP analysis indicate that local methylation of CYP7B1 promoter region is an important mechanism involved in down-regulation of CYP7B1 in human prostate tissue. CONCLUSIONS: This is the first report showing that CYP7B1 is overexpressed in high-grade PIN and in prostate cancer and that local methylation of CYP7B1 promoter region may have significant effect on gene transcription.


Subject(s)
Adenocarcinoma/enzymology , Cytochrome P-450 Enzyme System/biosynthesis , Estrogen Receptor beta/biosynthesis , Prostatic Neoplasms/enzymology , Steroid Hydroxylases/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aged , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 7 , DNA Methylation , Decitabine , Enzyme Inhibitors/pharmacology , Estrogen Receptor beta/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Male , Middle Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Steroid Hydroxylases/genetics
14.
J Steroid Biochem Mol Biol ; 104(3-5): 326-33, 2007 May.
Article in English | MEDLINE | ID: mdl-17467270

ABSTRACT

Dehydroepiandrosterone (DHEA) is 7alpha-hydroxylated by the cytochome P450 7B1 (CYP7B1) in the human brain and liver. This produces 7alpha-hydroxy-DHEA that is a substrate for 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) which exists in the same tissues and carries out the inter-conversion of 7alpha- and 7beta-hydroxy-DHEA through a 7-oxo-intermediary. Since the role of 11beta-HSD1 is to transform the inactive cortisone into active cortisol, its competitive inhibition by 7alpha-hydroxy-DHEA may support the paradigm of native anti-glucocorticoid arising from DHEA. Therefore, our objective was to use human tissues to assess the presences of both CYP7B1 and 11beta-HSD1. Human skin was selected then and used to test its ability to produce 7alpha-hydroxy-DHEA, and to test the interference of 7alpha- and 7beta-hydroxy-DHEA and 7-oxo-DHEA with the 11beta-HSD1-mediated oxidoreduction of cortisol and cortisone. Immuno-histochemical studies showed the presence of both CYP7B1 and 11beta-HSD1 in the liver, skin and tonsils. DHEA was readily 7alpha-hydroxylated when incubated using skin slices. A S9 fraction of dermal homogenates containing the 11beta-HSD1 carried out the oxidoreduction of cortisol and cortisone. Inhibition of the cortisol oxidation by 7alpha-hydroxy-DHEA and 7beta-hydroxy-DHEA was competitive with a Ki at 1.85+/-0.495 and 0.255+/-0.005 microM, respectively. Inhibition of cortisone reduction by 7-oxo-DHEA was of a mixed type with a Ki at 1.13+/-0.15 microM. These findings may support the previously proposed native anti-glucocorticoid paradigm and suggest that the 7alpha-hydroxy-DHEA production is a key for the fine tuning of glucocorticoid levels in tissues.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dehydroepiandrosterone/metabolism , Steroid Hydroxylases/metabolism , Cortisone/metabolism , Cytochrome P450 Family 7 , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/pharmacology , Female , Humans , Hydrocortisone/metabolism , Hydroxylation , Immunohistochemistry , In Vitro Techniques , Male , Oxidation-Reduction , Skin/enzymology , Skin/metabolism
15.
Drug Metabol Drug Interact ; 22(1): 1-23, 2006.
Article in English | MEDLINE | ID: mdl-17152345

ABSTRACT

Dehydroepiandrosterone (DHEA) is 7alpha-hydroxylated by the cytochrome P4507B1 in the liver, skin and brain, which are targets for glucocorticoids. 7alpha-Hydroxy-DHEA produced anti-glucocorticoid effects in vivo but the interference between the glucocorticoid hormone binding with its receptor could not be determined. In the organs mentioned above, circulating inactive cortisone is reduced to active cortisol by 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). 7alpha-Hydroxy-DHEA is also a substrate for this enzyme. Studies of 11beta-HSD1 action on 7alpha-hydroxy-DHEA show the reversible production of 7beta-hydroxy-DHEA through an intermediary 7-oxo-DHEA. Both the production of 7alpha-hydroxysteroids and their interference with the activation of cortisone into cortisol are basic to the concept of native anti-glucocorticoids. The cytochrome P4507B1 responsible for 7alpha-hydroxy-DHEA production and 11beta-HSD1 are key enzymes for the modulation of glucocorticoid action in humans. This is a promising new area for research.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Dehydroepiandrosterone/metabolism , Liver/metabolism , Steroid Hydroxylases/metabolism , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Bile Acids and Salts/metabolism , Cytochrome P450 Family 7 , Dehydroepiandrosterone/chemistry , Drug Interactions , Humans , Intestinal Mucosa/metabolism , Microsomes , Substrate Specificity , Yeasts/metabolism
16.
J Steroid Biochem Mol Biol ; 100(1-3): 95-105, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16713254

ABSTRACT

Circulating 3beta-hydroxysteroids including dehydroepiandrosterone (DHEA) are 7alpha-hydroxylated by the cytochrome P450-7B1 in the liver, skin and brain, which are the target organs of glucocorticoids. Anti-glucocorticoid effects with 7alpha-hydroxy-DHEA were observed in vivo without an interference with glucocorticoid binding to its receptor. In the organs mentioned above, the circulating inactive cortisone was reduced into active cortisol by the 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). We demonstrated that 7alpha-hydroxy-DHEA was also a substrate for this enzyme. Studies of the 11beta-HSD1 action on 7alpha-hydroxy-DHEA showed the reversible production of 7beta-hydroxy-DHEA through an intermediary 7-oxo-DHEA, and the kinetic parameters favored this production over that of active glucocorticoids. Both the production of 7alpha-hydroxysteroids and their interference with the activation of cortisone into cortisol are basic to the concept of native anti-glucocorticoids efficient at their production site. This opens a promising new area for research.


Subject(s)
Dehydroepiandrosterone/metabolism , Glucocorticoids/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Aging/metabolism , Animals , Brain/metabolism , Cortisone/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 7 , Humans , Hydrocortisone/metabolism , Kinetics , Liver/metabolism , Models, Biological , Receptors, Glucocorticoid/metabolism , Skin/metabolism , Steroid Hydroxylases/metabolism , Substrate Specificity
17.
J Steroid Biochem Mol Biol ; 99(4-5): 215-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16603347

ABSTRACT

The dehydroepiandrosterone (DHEA) 7alpha-hydroxylation in humans takes place in the liver, skin, and brain. These organs are targets for the glucocorticoid hormones where 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) activates cortisone through its reduction into cortisol. The putative interference of 7alpha-hydroxy-DHEA with the 11beta-HSD1-catalyzed reduction of cortisone into cortisol has been confirmed in preliminary works with human liver tissue preparations of the enzyme demonstrating the transformation of 7alpha-hydroxy-DHEA into 7-oxo-DHEA and 7beta-hydroxy-DHEA. However, the large production of 7beta-hydroxy-DHEA could not be explained satisfactorily. Therefore our objective was to study the role in the metabolism of oxygenated DHEA by recombinant human 11beta-HSD1 expressed in yeast. The 7alpha- and 7beta-hydroxy-DHEA were each oxidized into 7-oxo-DHEA with quite dissimilar K(M) (70 and 9.5 microM, respectively) but at equivalent V(max). In contrast, the 11beta-HSD1-mediated reduction of 7-oxo-DHEA led to the production of both 7alpha- and 7beta-hydroxy-DHEA with equivalent K(M) (1.1 microM) but with a 7beta-hydroxy-DHEA production characterized by a significantly greater V(max). The 7alpha-hydroxy-DHEA produced by the cytochrome CYP7B1 in tissues may exert anti-glucocorticoid effects through interference with the 11beta-HSD1-mediated cortisone reduction.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Dehydroepiandrosterone/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Base Sequence , Chromatography, Thin Layer , Cortisone/metabolism , DNA Primers , Genetic Vectors , Humans , Hydrocortisone/metabolism , Kinetics , Substrate Specificity
18.
Steroids ; 71(3): 240-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16371229

ABSTRACT

In this study the anti-oxidant effect of DHEA and 7alpha-hydroxy-DHEA against oxidative stress induced by colitis was investigated in vivo in rats. The two steroids were intraperitoneally injected once daily (50 mg/kg body weight) for 7 days before the induction of colitis that was effected by a daily treatment of 5% (w/v) dextran sodium sulfate (DSS) in drinking water for 7 days. This was quantified by the evidence of weight loss, rectal bleeding, increased wall thickness, and colon length. The inflammatory response was assessed by neutrophil infiltration after a histological examination and myeloperoxidase (MPO) activity measurement. Two markers of oxidative damage were measured in colon homogenates after the onset of DSS treatment: protein carbonyls and thiobarbituric acid-reacting substances. The colonic metabolism of corticosterone by 11beta-hydroxysteroid dehydrogenases types 1 and 2 (11beta-HSD) was investigated in control and treated animals. Results indicated that colitis caused a decrease in body weight and colon length. Severe lesions were observed in the colon with a reduced number of goblet cells which contained less mucins. The lesions were associated with increased MPO activity and oxidative damage. Colonic inflammation down and up regulated the 11beta-HSD2 and 11beta-HSD1, respectively. Treatments by DHEA and 7alpha-hydroxy-DHEA attenuated the inflammatory response when MPO activity decreased; but this did not increase the colonic oxidation of corticosterone into 11-dehydrocorticosterone. Both DHEA and 7alpha-hydroxy-DHEA exerted a significant anti-oxidant effect against oxidative stress induced by colitis through reducing the oxidative damage to proteins and lipids. This resulted in a moderate increase in the amount of colonic mucus. Both DHEA and 7alpha-hydroxy-DHEA may prove useful in the prevention or treatment of colitis.


Subject(s)
Colitis/drug therapy , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/therapeutic use , Dextran Sulfate/toxicity , Oxidative Stress/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Adjuvants, Immunologic/therapeutic use , Animals , Antioxidants/therapeutic use , Colitis/chemically induced , Colitis/metabolism , Male , Peroxidase/drug effects , Peroxidase/metabolism , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism , Weight Loss/drug effects
19.
Steroids ; 70(4): 319-26, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784286

ABSTRACT

Both dehydroepiandrosterone (DHEA) and epiandrosterone (EpiA) are substrate for cytochrome P450 species and enzymes that produce 7alpha- and 7beta-hydroxylated metabolites in the brain and other organs. In contrast to DHEA and EpiA, the 7-hydroxylated derivatives were shown to mediate neuroprotection, and 7beta-hydroxy-EpiA was the most potent. The suggested use of any of these steroids as drugs administered per os for neuroprotection requires the assessment of their metabolism in the human intestine and liver. To achieve this, we produced radio-labeled 7alpha-hydroxy-DHEA, 7beta-hydroxy-DHEA, 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA that were used as substrates in incubations with human intestine microsomes supplemented with reduced or oxidized cofactors. Identity of the radio-labeled metabolites obtained was determined by gas chromatography/mass spectrometry after comparison with authentic steroid references. The proportions of metabolites produced resulted from their radioactivity contents. The only metabolite obtained with DHEA, EpiA, 7alpha-hydroxy-DHEA and 7beta-hydroxy-DHEA substrates was its 17beta-reduced derivative, thus inferring the presence of 17beta-hydroxysteroid oxidoreductases in the human intestine microsomes. In addition to the 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA substrates, their 17beta-reduced metabolites were obtained with 7beta-hydroxy-EpiA and 7alpha-hydroxy-EpiA, respectively. The identity of the enzyme responsible for the 7alpha-hydroxy-EpiA/7beta-hydroxy-EpiA inter-conversion is unknown. The incubation conditions used produced these metabolites in low but significant yields that suggest their presence in the portal blood before access to the liver.


Subject(s)
Androsterone/metabolism , Dehydroepiandrosterone/metabolism , Intestinal Mucosa/metabolism , Microsomes/metabolism , Carbon Radioisotopes/metabolism , Gas Chromatography-Mass Spectrometry , Humans , Hydroxylation , Microsomes, Liver/metabolism , Reference Standards , Substrate Specificity
20.
Drug Metab Dispos ; 33(4): 563-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15650074

ABSTRACT

Dehydroepiandrosterone (DHEA) and 3beta-hydroxy-5alpha-androstan-17-one (epiandrosterone, EpiA) are both precursors for 7alpha- and 7beta-hydroxylated metabolites in the human brain. These 7-hydroxylated derivatives were shown to exert anti-glucocorticoid and neuroprotective effects. When these steroids are administered per os to humans, the first organ encountered is the liver, where extensive metabolism takes place. The objective of this work was to assess the cofactor dependence and metabolism of DHEA, EpiA, and their 7-hydroxylated derivatives in S9 fractions of human liver, using a radiolabeled steroid substrate for quantification and gas chromatography-mass spectrometry for identification. The best transformation yields were obtained with NADPH and were larger in female than in male. Results showed that both DHEA and EpiA mainly transformed into their 17beta-hydroxylated derivatives, 7- or 16alpha-hydroxylated metabolites under NAD(P)H conditions, and 5alpha-androstane-3,17-dione for EpiA under NAD(P)+ conditions. In turn, 7alpha-hydroxy-DHEA and 7beta-hydroxy-DHEA were partly transformed into each other via a 7-oxo-DHEA intermediate and were reduced into the 17beta-hydroxy derivative, respectively. The same type of transformations occurred for 7alpha-hydroxy-EpiA and 7beta-hydroxy-EpiA, except that no 7-oxo-EpiA intermediate was obtained. These findings determine the presence of enzymes responsible for the 7alpha- and 16alpha-hydroxylation in the human liver, the 11beta-hydroxysteroid dehydrogenase type 1 responsible for the oxidoreduction of the 7-hydroxylated substrates, and the 17beta-hydroxysteroid dehydrogenase responsible for the reduction of 17-oxo-steroids into 17beta-hydroxysteroids.


Subject(s)
Androsterone/metabolism , Dehydroepiandrosterone/metabolism , Liver/metabolism , Female , Humans , Hydroxylation , In Vitro Techniques , Male , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...