Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Front Neuroendocrinol ; 57: 100836, 2020 04.
Article in English | MEDLINE | ID: mdl-32217094

ABSTRACT

The enzymatic complex 5α-reductase (5α-R) and 3α/3ß-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Cholestenone 5 alpha-Reductase/physiology , Progesterone/metabolism , Testosterone/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Brain/enzymology , Cholestenone 5 alpha-Reductase/genetics , Female , Gene Expression , Humans , Male , Mental Disorders/enzymology , Neurodegenerative Diseases/enzymology , Neuroprotective Agents , Sex Characteristics
2.
J Exp Med ; 197(10): 1269-78, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12756265

ABSTRACT

The 3beta-hydroxysteroid dehydrogenase (3beta-HSD) isoenzymes play a key role in cellular steroid hormone synthesis. Vaccinia virus (VV) also synthesizes steroid hormones with a 3beta-HSD enzyme (v3beta-HSD) encoded by gene A44L. Here we examined the effects of v3beta-HSD in VV disease using wild-type (vA44L), deletion (vDeltaA44L), and revertant (vA44L-rev) viruses in a murine intranasal model. Loss of A44L was associated with an attenuated phenotype. Early (days 1-3) after infection with vDeltaA44L or control viruses the only difference observed between groups was the reduced corticosterone level in lungs and plasma of vDeltaA44L-infected animals. Other parameters examined (body weight, signs of illness, temperature, virus titres, the pulmonary inflammatory infiltrate, and interferon [IFN]-gamma levels) were indistinguishable between groups. Subsequently, vDeltaA44L-infected animals had reduced weight loss and signs of illness, and displayed a vigorous pulmonary inflammatory response. This was characterized by rapid recruitment of CD4+ and CD8+ lymphocytes, enhanced IFN-gamma production and augmented cytotoxic T lymphocyte activity. These data suggest that steroid production by v3beta-HSD contributes to virus virulence by inhibiting an effective inflammatory response to infection.


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Glucocorticoids/physiology , Immune Tolerance , Vaccinia virus/enzymology , Vaccinia/immunology , Viral Proteins/physiology , Animals , Corticosterone/blood , Cytotoxicity, Immunologic , Female , Inflammation/prevention & control , Interferon-gamma/biosynthesis , Lung/immunology , Lung/virology , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Vaccinia virus/pathogenicity , Virulence , Virus Replication
3.
Toxicol Lett ; 181(1): 1-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18616992

ABSTRACT

Resistance towards anticancer drugs is a general problem upon chemotherapy. Among the mechanisms of resistance, metabolic inactivation by carbonyl reduction is a major cause of chemotherapy failure that applies to drugs bearing a carbonyl moiety. Oracin is a promising anticancer drug which is presently in phase II clinical trials. Pharmacokinetic studies have revealed that oracin undergoes metabolic inactivation by carbonyl reduction. In the present study, we provide evidence that AKR1C3, a member of the aldo-keto reductase (AKR) superfamily, catalyzes the inactivation of oracin. Moreover, AKR1C3 does also mediate C13 carbonyl reduction of doxorubicin to its inactive hydroxy metabolite doxorubicinol. Doxorubicinol, however, has also been considered responsible for the cardiomyopathy observed upon doxorubicin chemotherapy. Since AKR1C3 is overexpressed in hormone-dependent malignancies like prostate and breast cancer, coadministration of AKR1C3 inhibitors might enhance the chemotherapeutic efficacy of oracin and doxorubicin, and simultaneously reduce the risk of cardiomyopathy upon doxorubicin treatment.


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Antibiotics, Antineoplastic/metabolism , Doxorubicin/metabolism , Ethanolamines/metabolism , Hydroxyprostaglandin Dehydrogenases/physiology , Isoquinolines/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3 , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification
4.
Mol Cell Endocrinol ; 248(1-2): 182-91, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16417966

ABSTRACT

Human aldo-keto reductases (AKR) of the 1A, 1B, 1C and 1D subfamilies are involved in the pre-receptor regulation of nuclear (steroid hormone and orphan) receptors by regulating the local concentrations of their lipophilic ligands. AKR1C3 is one of the most interesting isoforms. It was cloned from human prostate and the recombinant protein was found to function as a 3-, 17- and 20-ketosteroid reductase with a preference for the conversion of Delta4-androstene-3,17-dione to testosterone implicating this enzyme in the local production of active androgens within the prostate. Using a validated isoform specific real-time RT-PCR procedure the AKR1C3 transcript was shown to be more abundant in primary cultures of epithelial cells than stromal cells, and its expression in stromal cells increased with benign and malignant disease. Using a validated isoform specific monoclonal Ab, AKR1C3 protein expression was also detected in prostate epithelial cells by immunoblot analysis. Immunohistochemical staining of prostate tissue showed that AKR1C3 was expressed in adenocarcinoma and surprisingly high expression was observed in the endothelial cells. These cells are a rich source of prostaglandin G/H synthase 2 (COX-2) and vasoactive prostaglandins (PG) and thus the ability of recombinant AKR1C enzymes to act as PGF synthases was compared. AKR1C3 had the highest catalytic efficiency (kcat/Km) for the 11-ketoreduction of PGD2 to yield 9alpha,11beta-PGF2 raising the prospect that AKR1C3 may govern ligand access to peroxisome proliferator activated receptor (PPARgamma). Activation of PPARgamma is often a pro-apoptotic signal and/or leads to terminal differentiation, while 9alpha,11beta-PGF2 is a pro-proliferative signal. AKR1C3 is potently inhibited by non-steroidal anti-inflammatory drugs suggesting that the cancer chemopreventive properties of these agents may be mediated either by inhibition of AKR1C3 or COX. To discriminate between these effects we developed potent AKR1C inhibitors based on N-phenylanthranilic acids that do not inhibit COX-1 or COX-2. These compounds can now be used to determine the role of AKR1C3 in producing two proliferative signals in the prostate namely testosterone and 9alpha,11beta-PGF2.


Subject(s)
3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Prostatic Diseases/enzymology , 3-Hydroxysteroid Dehydrogenases/analysis , 3-Hydroxysteroid Dehydrogenases/physiology , Aldo-Keto Reductase Family 1 Member C3 , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Dinoprost/biosynthesis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Gonadal Steroid Hormones/biosynthesis , Gonadal Steroid Hormones/metabolism , Humans , Hydroxyprostaglandin Dehydrogenases/analysis , Hydroxyprostaglandin Dehydrogenases/physiology , Male , Prostate/enzymology , Prostatic Diseases/genetics , Structure-Activity Relationship , Testosterone/biosynthesis , Transcription, Genetic
5.
Cancer Res ; 55(18): 4105-13, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7664287

ABSTRACT

Rat liver 3 alpha-hydroxysteroid/dihydrodiol dehydrogenase (3 alpha-HSD/DD) is a member of the aldo-keto reductase gene superfamily. It displays high constitutive expression and inactivates circulating steroid hormones and suppresses the formation of polycyclic aromatic hydrocarbon anti- and syn-diol-epoxides (ultimate carcinogens). To elucidate mechanisms responsible for constitutive expression of the 3 alpha-HSD/DD gene a rat genomic library obtained from adult Sprague-Dawley female liver (HaeIII partial digest) was screened, using a probe corresponding to the 5'-end of the cDNA (-15 to +250), and a 15.8-kb genomic clone was isolated. Sequencing revealed that 6.3 kb contained exon 1 (+16 to +138 bp) plus additional introns and exons. The transcription start site (+1) was located by primer extension analysis, and the initiation codon, ATG, was located at +55 bp. The remaining 9.5 kb represented the 5'-flanking region of the rat 3 alpha-HSD/DD gene. A 1.6-kb fragment of this region was sequenced. A TATTTAA sequence (TATA box) was found at 33 bp upstream from the major transcription start site. cis-acting elements responsible for the constitutive expression of the rat 3 alpha-HSD/DD gene were located on the 5'-flanking region by transient transfection of reporter-gene (chloramphenicol acetyl transferase, CAT) constructs into human hepatoma cells (HepG2). CAT assays identified the basal promoter between (-199 and +55 bp), the presence of a proximal enhancer (-498 to -199 bp) which stimulated CAT activity 6-fold, the existence of a powerful silencer (-755 to -498 bp), and a strong distal enhancer (-4.0 to -2.0 kb) which increased CAT activity by 20-40-fold. A computer search of available consensus sequences for trans-acting factors revealed that a cluster of Oct-sites were uniquely located in the silencer region. Using the negative response element (-797 to -498 bp) as a probe and nuclear extracts from HepG2 cells, three bands were identified by gel mobility shift assay, indicating the presence of protein binding sites in this proposed negative response element. All three bands were supershifted with anti-Oct-1 mAb, suggesting that Oct-1 may be the repressor. The 5'-flanking region also contained an AP-1 site, an estrogen response element, and a glucocorticoid response element, which together may comprise a steroid response unit.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
3-Hydroxysteroid Dehydrogenases/genetics , Oxidoreductases/genetics , 3-Hydroxysteroid Dehydrogenases/physiology , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) , Amino Acid Sequence , Animals , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Cloning, Molecular , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Enzymologic , Host Cell Factor C1 , Molecular Sequence Data , Octamer Transcription Factor-1 , Oxidoreductases/physiology , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
6.
Reprod Biol Endocrinol ; 3: 11, 2005 Apr 03.
Article in English | MEDLINE | ID: mdl-15804366

ABSTRACT

In mouse ovaries, the enzyme 3 beta-hydroxysteroid dehydrogenase (HSD) is distributed between microsomes and mitochondria. Throughout the follicular phase of the estrous cycle, the HSD activity in microsomes is predominant; whereas, after LH stimulation, HSD activity during the luteal phase is highest in the mitochondria. The current study examined whether or not LH stimulation always results in an increase in mitochondrial HSD activity. This was accomplished by measuring the HSD activity in microsomal and mitochondrial fractions from ovaries of pregnant mice. These animals have two peaks of LH during gestation, and one peak of LH after parturition. It was found that mitochondrial HSD activity was highest after each peak of LH. It is proposed that mitochondrial HSD is essential for the synthesis of high levels of progesterone. The increase in HSD activity in mitochondria after LH stimulation occurs because: 1) LH initiates the simultaneous synthesis of HSD and the cholesterol side-chain cleavage enzyme (P450scc); and, 2) HSD and P450scc bind together to form a complex, which becomes inserted into the inner membrane of the mitochondria. High levels of progesterone are synthesized by mitochondrial HSD because: 1) the requisite NAD+ cofactor for progesterone synthesis is provided directly by the mitochondria, rather than indirectly via the rate limiting malate-aspartate shuttle; and, 2) the end-product inhibition of P450scc by pregnenolone is eliminated because pregnenolone is converted to progesterone.


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Corpus Luteum/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/physiology , Progesterone/biosynthesis , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Electron Transport Complex IV/metabolism , Female , Luteinizing Hormone/metabolism , Mice , Microsomes/enzymology , Mitochondrial Proteins/metabolism , Models, Biological , Pregnancy
7.
Front Biosci ; 4: D596-600, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10417059

ABSTRACT

Prostate cancer will be diagnosed in about 179,300 men in the US in 1999 alone. Some 37,000 individuals die of this disease annually. Prostate cancer is characterized by a substantial racial/ethnic variation in risk: highest in African-American men, lowest in Asian men and intermediate in Caucasian and Latino men. We set out to investigate as our central hypothesis that genetic variants of genes involved in androgen metabolism by themselves and in combination significantly contribute to prostate cancer progression and its racial/ethnic variation. Specifically, we examined the hypothesis that DNA sequence (allelic) variations in the type II (or prostatic) steroid 5alpha-reductase (SRD5A2) gene contribute substantially to the risk and progression of prostate cancer particularly across racial/ethnic lines. The "candidate gene", SRD5A2, was chosen because the reaction product [i.e. dihydrotestosterone (DHT)] of the enzyme encoded by this gene modulates directly cell division in the prostate. DHT binds to the androgen receptor (AR) and the DHT-AR complex leads to the transactivation of a variety of genes which ultimately modulates cell division in the prostate. Epidemiologic evidence suggests that variation in DHT levels play an important role in risk of prostate cancer. Thus, steroid 5alpha-reductase activity encoded by SRD5A2 variant alleles may be important in regulating intraprostatic DHT steady state levels by controlling its biosynthesis. A second candidate gene, the type II 3beta-hydroxysteroid dehydrogenase (HSD3B2) gene, encodes the enzyme that initiates the metabolic inactivation of testosterone (T) to DHT. We have identified allelic variants in this gene as well. Here I review our strategy for identifying candidate genes for prostate cancer, a multifactorial disease. I summarize the significant findings, particularly of allelic variants in the HSD3B2 and SRD5A2 genes and discuss how they by themselves, in combination and through interactions with the environment may play a role in prostate cancer predisposition and its progression. Our approach, a multidisciplinary genomic genetic (GEN GEN) attack on the problem, may be useful in the analysis of other complex phenotypes as well.


Subject(s)
Prostatic Neoplasms/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/physiology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , Androgens/physiology , Disease Progression , Genetic Predisposition to Disease , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism
8.
J Endocrinol ; 129(2): 233-43, 1991 May.
Article in English | MEDLINE | ID: mdl-2040858

ABSTRACT

We have previously suggested that in porcine granulosa cells, a putative intermediate, 5(10)-oestrene-3,17-dione is involved in 4-oestrene-3,17-dione (19-norandrostenedione; 19-norA) and 4-oestren-17 beta-ol-3-one (19-nortestosterone: 19-norT) formation from C19 aromatizable androgens. In this study, luteal cells prepared from porcine, bovine and rat corpora lutea by centrifugal elutriation were used as a source of 3 beta-hydroxysteroid dehydrogenase/isomerase in order to investigate the role of this enzyme in the biosynthesis of 19-norsteroids. Small porcine luteal cells made mainly 19-norT and large porcine luteal cells 19-norA from 5(10)-oestrene-3 beta,17 beta-diol, the reduced product of the putative intermediate 5(10)-oestrene-3,17-dione. However, neither small nor large cells metabolized androstenedione to 19-norsteroids. Serum and serum plus LH significantly stimulated formation of both 19-norA and 19-norT from 5(10)-oestrene-3 beta,17 beta-diol, compared with controls. Inhibitors of the 3 beta-hydroxysteroid dehydrogenase/isomerase (trilostane and cyanoketone) significantly reduced formation of 19-norT in small porcine luteal cells and 19-norA in large porcine luteal cells, although they were effective at different concentrations in each cell type. In parallel incubations, formation of [4-14C]androstenedione from added [4-14C]dehydroepiandrosterone was also inhibited by cyanoketone in both small and large porcine luteal cells in a dose-dependent manner; however, trilostane (up to 100 mumol/l) did not inhibit androstenedione formation in large porcine luteal cells. In addition, the decrease in progesterone synthesis induced by trilostane and cyanoketone (100 mumol/l each) was accompanied by a parallel accumulation of pregnenolone in both cell types. These results suggest that 3 beta-hydroxysteroid dehydrogenase/isomerase, or a closely related enzyme, present in small and large porcine luteal cells can convert added 5(10)-3 beta-hydroxysteroids into 19-nor-4(5)-3-ketosteroids in vitro. In the porcine ovarian follicle, therefore, formation of 19-norA from androstenedione can be envisaged as a two-step enzymatic process: 19-demethylation of androstenedione to produce the putative intermediate 5(10)-oestrene-3,17-dione, and subsequent isomerization to 19-norA. In contrast to granulosa cells, porcine luteal cells synthesized 19-norA or 19-norT only when provided with the appropriate substrate. Unfractionated rat luteal cells also metabolized 5(10)-oestrene-3 beta,17 beta-diol to a mixture of 19-norA and 19-norT; conversion was inhibited by trilostane. In addition, small bovine luteal cells synthesized mainly 19-norT and formation was also inhibited by trilostane and cyanoketone.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Estrenes/metabolism , Luteal Cells/metabolism , Norsteroids/metabolism , Androstenedione/analogs & derivatives , Androstenedione/biosynthesis , Animals , Cattle , Cells, Cultured , Chromatography, High Pressure Liquid , Female , Nandrolone/metabolism , Radioimmunoassay , Rats , Swine
9.
Placenta ; 15(1): 79-88, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8208672

ABSTRACT

An initial group of term (36-41 6/7 weeks), preterm (less than 36 weeks), and post-term (42 or more weeks) placentae were collected from women at delivery to determine the placental levels of important steroids and steroidogenic enzymes involved in the oestrogen synthesis pathway as a function of gestational age. A second group of placentae were obtained from women delivering at term before and after the onset of labour. Placentae were evaluated individually for cytosolic steroid hormone levels and microsomal steroidogenic enzyme activities. Oestradiol (E2), oestrone (E1), progesterone (P), and delta-4-androstenedione (A) were measured by radioimmunoassay in placental cytosols. Aromatase (AR), sulphatase (S), and 3 beta-hydroxysteroid dehydrogenase/isomerase (3 beta HSD) activities were assayed in placental microsomes. Cytosolic concentrations of E1, E2, P, and A did not differ with respect to gestational age. Correspondingly, the microsomal enzyme activities of 3 beta HSD, S, and AR did not vary as a function of gestational age. However, when patients at term who were in labour prior to delivery were compared to those who were not, the placental cytosolic level of E1 was found to be threefold higher in the non-labouring group (4572 versus 1427 pg/mg cytosolic protein, P < 0.025). Additionally, microsomal aromatase activity was also significantly higher in the non-labouring patients (46 versus 19 pM/min/mg protein, P < 0.025), while the E2 to P ratio in the labouring patients was twice that of the non-labouring group, a difference which was significant at the P < 0.025 level (Wilcoxon rank sum test). These data suggest that at term, prior to labour, the placental production of E1 by AR is high, and that AR activity and E1 levels fall significantly after the onset of labour. Also, the placental cytosolic concentration of the more active oestrogen, E2, demonstrates stable to rising levels with a significant increase in E2/P after the onset of labour. We theorize that in the term pregnancy prior to labour, E1 may represent a large but relatively inactive intracellular oestrogen pool which is maintained by high AR activity, and may function to protect the pregnant local uterine environment from the more oxytocic effects of E2.


Subject(s)
Estrogens/physiology , Labor, Obstetric/physiology , Placenta/physiology , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/physiology , Androstenedione/analysis , Androstenedione/physiology , Aromatase/metabolism , Aromatase/physiology , Estradiol/analysis , Estradiol/physiology , Estrogens/metabolism , Female , Humans , Placenta/chemistry , Placenta/metabolism , Pregnancy , Progesterone/analysis , Progesterone/physiology , Sulfatases/metabolism , Sulfatases/physiology
10.
Mol Cell Endocrinol ; 186(2): 137-41, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11900886

ABSTRACT

The human corpus luteum (CL) undergoes a dynamic cycle of differentiation, steroid hormone production and regression during the course of non-fertile cycles. In humans and other primates, luteal steroidogenesis is absolutely dependent on pituitary-derived LH. However, changes in LH and LH receptor expression do not explain the marked decline in progesterone production at the end of the luteal phase. Changes in the level of the steroidogenic acute regulatory protein (StAR), a gene whose expression is controlled by LH most likely account for the cyclic pattern of progesterone production. During the mid-to-late luteal phase of a fertile cycle, chorionic gonadotropin (hCG) rescues the CL, overcoming the actions of the factors inducing luteolysis. Although the agents causing regression of the CL in a non-fertile cycle are not yet known, intra-luteal growth factors and cytokines that modify the action of LH probably contribute to the reduction of StAR expression and the subsequent fall in progesterone production.


Subject(s)
Corpus Luteum/metabolism , Luteal Phase/physiology , Progesterone/biosynthesis , 3-Hydroxysteroid Dehydrogenases/physiology , Animals , Cholesterol/metabolism , Cholesterol Side-Chain Cleavage Enzyme/physiology , Chorionic Gonadotropin/physiology , Corpus Luteum/cytology , Corpus Luteum Maintenance/physiology , Female , Gene Expression Regulation , Granulosa Cells/metabolism , Humans , Luteinizing Hormone/physiology , Phosphoproteins/genetics , Phosphoproteins/physiology , Pregnancy , Primates/metabolism , Receptors, LH/physiology
11.
Fertil Steril ; 26(12): 1198-202, 1975 Dec.
Article in English | MEDLINE | ID: mdl-803034

ABSTRACT

Immunization of female rats with crude preparations of delta 5-3 beta-hydroxysteroid dehydrogenase combined with complete Freund's adjuvant resulted in suppression of the endogenous enzyme activity in the ovarian interstitial tissue and the corpora lutea. As a result, estrous cyclicity, nidation, or maintenance of pregnancy was adversely affected in proportion to the level of the antibody to the enzyme following the series of immunizations. On the basis of the biologic response, it can be assumed that progesterone synthesis was reduced as a result of neutralization of enzyme action in the ovary by the formation of antibodies to the delta 5-3 beta-hydroxysteroid dehydrogenase. Further studies are required to verify this assumption.


Subject(s)
3-Hydroxysteroid Dehydrogenases/physiology , Immunization , Animals , Corpus Luteum/immunology , Female , Ovary/immunology , Rats
12.
Steroids ; 62(1): 101-11, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9029723

ABSTRACT

Mammalian 3 alpha-hydroxysteroid dehydrogenases (3 alpha-HSDs) inactivate circulating steroid hormones, and in target tissues regulate the occupancy of steroid hormone receptors. Molecular cloning indicates that 3 alpha-HSDs are members of the aldo-keto reductase (AKR) superfamily and display high sequence identity (> 60%). Of these, the most extensively characterized is rat liver 3 alpha-HSD. X-ray crystal structures of the apoenzyme and the E.NADP+ complex have been determined and serve as structural templates for other 3 alpha-HSDs. These structures reveal that rat liver 3 alpha-HSD adopts an (alpha/beta)8-barrel protein fold. NAD(P)(H) lies perpendicular to the barrel axis in an extended conformation, with the nicotinamide ring at the core of the barrel, and the adenine ring at the periphery of the structure. The nicotinamide ring is stabilized by interaction with Y216, S166, D167, and Q190, so that the A-face points into the vacant active site. The 4-pro-(R) hydrogen transferred in the oxidoreduction of steroids is in close proximity to a catalytic tetrad that consists of D50, Y55, K84, and H117. A water molecule is within hydrogen bond distance of H117 and Y55, and its position may mimic the position of the carbonyl of a 3-ketosteroid substrate. The catalytic tetrad is conserved in members of the AKR superfamily and resides at the base of an apolar cleft implicated in binding steroid hormone. The apolar cleft consists of a side of apolar residues (L54, W86, F128, and F129), and opposing this side is a flexible loop that contains W227. These constraints suggest that the alpha-face of the steroid would orient itself along that side of the cleft containing W86. Site-directed mutagenesis of the catalytic tetrad indicates that Y55 and K84 are essential for catalysis. Y55S and Y55F mutants are catalytically inactive, but still form binary (E.NADPH) and ternary (E.NADH.Testosterone) complexes; by contrast K84R and K84M mutants are catalytically inactive, but do not bind steroid hormone. The reliance on a Tyr/Lys pair is reminiscent of catalytic mechanisms proposed for other AKR members as well as for HSDs that belong to the short-chain dehydrogenase/reductase (SDR) family, in which Tyr is the general acid, with its pKa being lowered by Lys. Superimposition of the nicotinamide rings in the structures of 3 alpha-HSD (an AKR) and 3 alpha, 20 beta-HSD (an SDR) show that the Tyr/Lys pairs are positionally conserved, suggesting convergent evolution across protein families to a common mechanism for HSD catalysis. W86Y and W227Y mutants bind testosterone to the E.NADH complex, with effective increases in Kd of 8- and 20-fold. These data provide the first evidence that the side of the apolar cleft containing W86 and the opposing flexible loop containing W227 are parts of the steroid-binding site. Detailed mutagenesis studies of the apolar cleft and elucidation of a ternary complex structure will ultimately provide details of the determinants that govern steroid hormone recognition. These determinants could provide a rational basis for structure-based inhibitor design.


Subject(s)
3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/physiology , Alcohol Oxidoreductases/chemistry , Liver/enzymology , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) , Alcohol Oxidoreductases/physiology , Aldehyde Reductase , Aldo-Keto Reductases , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Rats , Sequence Homology, Amino Acid , Steroids/metabolism
13.
Folia Biol (Krakow) ; 48(1-2): 19-24, 2000.
Article in English | MEDLINE | ID: mdl-11080913

ABSTRACT

The study was designed to compare the direct effect of three prolactin-like hormones on steroidogenesis of ovine luteal cells collected at day 40-45 of pregnancy. 100 ng/ml of ovine placental lactogen or 100 ng/ml of ovine growth hormone or 100 ng/ml of ovine prolactin were added to the media of luteal cell cultures. After 48 h incubation, all cultures were terminated and the media were frozen until further steroid analysis. To determine to what extent growth hormone (GH), prolactin (PRL) and lactogen (PL) regulate the activity of 3 beta-HSD, an enzyme involved in progesterone synthesis, the classical steroidal competitive inhibitor of 3 beta-HSD trilostane, was investigated for its effects on basal and GH-, PRL-, and PL-stimulated progesterone biosynthesis since there is a possibility that the luteotropic effect of these hormones are mediated via 3 beta-HSD. oPL resulted in an increase of progesterone secretion in a statistically significant manner, while GH or PRL had no effect on progesterone secretion. A decrease in progesterone secretion as an effect of 100 mM trilostane was observed in all culture types. An explanation for the luteotropic effect of PL and the lack of this effect for GH is that the GH receptor associates with a different molecule within the ovarian tissue and forms a heterodimeric receptor for PL, and the possibility that physiological effects of native oPL may be mediated through its binding to specific PL receptors, which have low affinities for oGH and oPRL.


Subject(s)
Corpus Luteum/metabolism , Growth Hormone/physiology , Placental Lactogen/physiology , Progesterone/metabolism , Prolactin/physiology , Sheep/physiology , 3-Hydroxysteroid Dehydrogenases/physiology , Animals , Corpus Luteum/physiology , Female , Male , Pregnancy , Progesterone/analysis , Radioimmunoassay/veterinary , Statistics, Nonparametric
14.
Nihon Rinsho ; 62(2): 339-43, 2004 Feb.
Article in Japanese | MEDLINE | ID: mdl-14968542

ABSTRACT

Disorders of androgen biosynthesis are a relatively rare cause of sexual ambiguity in 46XY genetic males. The biosynthesis of androgen requires the steroidogenic acute regulatory protein (StAR) and the steroidogenic enzymes P450scc, 3 beta HSDII, P450c17, 17 beta HSDIII, and 5 alpha-reductase. Deficiencies have been reported in these enzymes, leading to male pseudohermaphroditism. Here we describe three enzymes 3 beta HSDII, P450c17, and 17 beta HSDIII, their roles and mechanisms of action special reference to diagnosis and treatment for the patients with deficiencies of these enzymes.


Subject(s)
Androgens/biosynthesis , Disorders of Sex Development/etiology , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/physiology , 3-Hydroxysteroid Dehydrogenases/deficiency , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/physiology , Disorders of Sex Development/diagnosis , Disorders of Sex Development/therapy , Humans , Male , Mutation , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/physiology
16.
Anim Reprod Sci ; 140(3-4): 138-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23896394

ABSTRACT

In the present paper, cellular composition of buffalo corpus luteum (CL) with its functional characterization based on 3ß-HSD and progesterone secretory ability at different stages of estrous cycle and pregnancy was studied. Buffalo uteri along with ovaries bearing CL were collected from the local slaughter house. These were classified into different stages of estrous cycle (Stage I, II, III and IV) and pregnancy (Stage I, II and III) based on morphological appearance of CL, surface follicles on the ovary and crown rump length of conceptus. Luteal cell population, progesterone content and steroidogenic properties were studied by dispersion of luteal cells using collagenase type I enzyme, RIA and 3ß-HSD activity, respectively. Large luteal cells (LLC) appeared as polyhedral or spherical in shape with a centrally placed large round nucleus and an abundance of cytoplasmic lipid droplets. However, small luteal cells (SLC) appeared to be spindle shaped with an eccentrically placed irregular nucleus and there was paucity of cytoplasmic lipid droplets. The size of SLC (range 12-23µm) and LLC (range 25-55µm) increased (P<0.01) with the advancement of stage of estrous cycle and pregnancy. The mean progesterone concentration per gram and per CL increased (P<0.01) from Stage I to III of estrous cycle with maximum concentration at Stage III of estrous cycle and pregnancy. The progesterone concentration decreased at Stage IV (day 17-20) of estrous cycle coinciding with CL regression. Total luteal cell number (LLC and SLC) also increased (P<0.01) from Stage I to III of estrous cycle and decreased (P<0.05), thereafter, at Stage IV indicating degeneration of luteal cells and regression of the CL. Total luteal cell population during pregnancy also increased (P<0.01) from Stage I to II and thereafter decreased (P>0.05) indicating cessation of mitosis. Increased (P<0.05) large luteal cell numbers from Stage I to III of estrous cycle and pregnancy coincided with the increased progesterone secretion and 3ß-HSD activity of CL. Thus, proportionate increases of large compared with small luteal cells were primarily responsible for increased progesterone secretion during the advanced stages of the estrous cycle and pregnancy. Total luteal cells and progesterone content per CL during the mid-luteal stage in buffalo as observed in the present study seem to be less than with cattle suggesting inherent luteal deficiency.


Subject(s)
Buffaloes/physiology , Corpus Luteum/physiology , Estrous Cycle/physiology , Ovary/physiology , 3-Hydroxysteroid Dehydrogenases/analysis , 3-Hydroxysteroid Dehydrogenases/physiology , Animals , Corpus Luteum/cytology , Corpus Luteum/enzymology , Female , Histocytochemistry/veterinary , Luteal Cells/cytology , Luteal Cells/enzymology , Luteal Cells/physiology , Ovary/cytology , Pregnancy , Progesterone/analysis , Progesterone/physiology
18.
Cancer Res ; 70(4): 1573-84, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145130

ABSTRACT

PR-104, currently in phase II clinical trials, is a phosphate ester pre-prodrug which is converted in vivo to its cognate alcohol, PR-104A, a prodrug designed to exploit tumor hypoxia. Bioactivation occurs via one-electron reduction to DNA crosslinking metabolites in the absence of oxygen. However, certain tumor cell lines activate PR-104A in the presence of oxygen, suggesting the existence of an aerobic nitroreductase. Microarray analysis identified a cluster of five aldo-keto reductase (AKR) family members whose expressions correlated with aerobic metabolism of PR-104A. Plasmid-based expression of candidate genes identified aldo-keto reductase 1C3 as a novel nitroreductase. AKR1C3 protein was detected by Western blot in 7 of 23 cell lines and correlated with oxic PR-104A metabolism, an activity which could be partially suppressed by Nrf2 RNAi knockdown (or induced by Keap1 RNAi), indicating regulation by the ARE pathway. AKR1C3 was unable to sensitize cells to 10 other bioreductive prodrugs and was associated with single-agent PR-104 activity across a panel of 9 human tumor xenograft models. Overexpression in two AKR1C3-negative tumor xenograft models strongly enhanced PR-104 antitumor activity. A population level survey of AKR1C3 expression in 2,490 individual cases across 19 cancer types using tissue microarrays revealed marked upregulation of AKR1C3 in a subset including hepatocellular, bladder, renal, gastric, and non-small cell lung carcinoma. A survey of normal tissue AKR1C3 expression suggests the potential for tumor-selective PR-104A activation by this mechanism. These findings have significant implications for the clinical development of PR-104.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Aerobiosis/physiology , Hydroxyprostaglandin Dehydrogenases/metabolism , Nitrogen Mustard Compounds/pharmacokinetics , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/physiology , Aldo-Keto Reductase Family 1 Member C3 , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/physiology , Inhibitory Concentration 50 , Mice , Mice, Nude , Models, Biological , Nitrogen Mustard Compounds/metabolism , Oxidation-Reduction/drug effects , Oxygen/pharmacology , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Tissue Array Analysis , Xenograft Model Antitumor Assays
20.
Endocrinology ; 150(4): 1748-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19036885

ABSTRACT

Adrenarche is thought to be experienced only by humans and some Old World primates despite observed regression of an adrenal fetal zone and establishment of a functional zona reticularis (ZR) in other species like rhesus macaques. Adrenal differentiation remains poorly defined biochemically in nonhuman primates. The present studies defined ZR development in the neonatal rhesus by examining androgen synthetic capacity and factors affecting it in rhesus and marmoset adrenals. Western immunoblots examined expression of 17alpha-hydroxylase/17,20-lyase cytochrome P450 (P450c17), cytochrome b5 (b5), and 3beta-hydroxysteroid dehydrogenase (3betaHSD), among other key enzymes. 17,20-lyase activity was quantified in adrenal microsomes, as was the contribution of b5 to 17,20-lyase activity in microsomes and cell transfection experiments with rhesus and marmoset P450c17. Expression of b5 increased from birth to 3 months, and was positively correlated with age and 17,20-lyase activity in the rhesus. Recombinant b5 addition stimulated 17,20-lyase activity to an extent inversely proportional to endogenous levels in adrenal microsomes. Although 3betaHSD expression also increased with age, P450c17, 21-hydroxylase cytochrome P450, and the redox partner, reduced nicotinamide adenine dinucleotide phosphate-cytochrome P450 oxidoreductase, did not; nor did recombinant cytochrome P450 oxidoreductase augment 17,20-lyase activity. Cotransfection with b5 induced a dose-dependent increase in dehydroepiandrosterone synthesis by both nonhuman primate P450c17 enzymes. We conclude that the increase in 17,20-lyase activity characteristic of an adrenarche in rhesus macaques is driven primarily by increased b5 expression, without the need for a decrease in 3betaHSD, as suggested from human studies. The rhesus macaque is a relevant and accessible model for human ZR development and adrenal function.


Subject(s)
Cytochromes b5/metabolism , Macaca mulatta/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/physiology , Animals , Animals, Newborn , Blotting, Western , Cell Line , Chromatography, Thin Layer , Cytochromes b5/genetics , Cytochromes b5/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Microsomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL