Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
J Cell Mol Med ; 25(14): 6773-6785, 2021 07.
Article En | MEDLINE | ID: mdl-34114342

Metabolism of progesterone (P4) by the enzyme 20α hydroxysteroid dehydrogenase (20α-HSD) in myometrial cells is postulated to be a mechanism for P4 withdrawal, which occurs concomitant to uterine inflammation (physiologic or infection-induced) and associated activation of transcription factors: NF-кB and AP-1, common to term and preterm labour. We found that 20α-HSD protein is significantly increased in human myometrium during term labour, and in mouse uterus during term and preterm labour. Treatment of human myometrial cells with the pro-inflammatory mediators, lipopolysaccharide (LPS, mimicking infection) and 12-O-tetradecanoylphorbol-13-acetate (TPA, mimicking inflammation), induced 20α-HSD gene expression and increased 20α-HSD protein abundance. LPS treatment decreased P4 release into the culture medium and resulted in up-regulation of GJA1 in the hTERT-HM cells. The NF-кB /AP-1 transcription factors mediated effects of LPS and TPA on 20α-HSD gene transcription. Both pro-inflammatory stimuli induced 20α-HSD promoter activity in LPS/TPA-treated cells which was significantly attenuated by inhibition of NF-кB (JSH: 20 µM) or AP-1 signalling (T5224: 10 µM). Deletion of NF-кB consensus sites abrogated LPS-mediated promoter induction, while removal of AP-1 sites reversed the TPA-mediated induction of 20α-HSD promoter. We conclude that inflammatory stimuli (physiologic or pathologic) that activate NF-кB or AP-1 induce 20α-HSD transcription and subsequent local P4 withdrawal resulting in up-regulation of GJA1 and activation of myometrium that precedes labour.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Myometrium/metabolism , NF-kappa B/metabolism , Premature Birth/metabolism , Progesterone/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Adult , Animals , Connexin 43/genetics , Connexin 43/metabolism , Female , HEK293 Cells , Humans , Mice , Myometrium/drug effects , NF-kappa B/genetics , Pregnancy , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
2.
Sci China Life Sci ; 61(8): 966-974, 2018 08.
Article En | MEDLINE | ID: mdl-29804163

To directly assess the molecular function of the monkey 20α-hydroxysteroid dehydrogenase (20α-HSD) promoter, we generated transgenic mice (tg) expressing enhanced green fluorescent protein (EGFP) under control of this promoter. We demonstrated that prostaglandin F2α induced 20α-HSD promoter activity in CHO cells in a dose-dependent manner. Furthermore, forskolin treatment markedly reduced 20α-HSD promoter activity, and prolactin exhibited weak inhibitory activity. The transgenic mouse obtained one positive founder male. The transgene was propagated in 10 successive generations without any notable defects to the progeny. EGFP and 20α-HSD in the tg mice were colocalized in the luteal cells of the ovary during late pregnancy. Strong EGFP and 20α-HSD protein signals were also detected in the adult testis. Immunohistochemical analysis revealed high EGFP levels in the seminiferous epithelium, whereas 20α-HSD was expressed in the seminiferous tubules. Our data suggest that the ovaries in monkey and mouse exhibit similar expression patterns of 20α-HSD during pregnancy. However, the expression pattern of EGFP in tg mice testis slightly differed from that of the endogenous 20α-HSD. Further investigation is required to elucidate the functional mechanisms underlying regulation of the monkey 20α-HSD promoter in the tg mice.


20-alpha-Hydroxysteroid Dehydrogenase/genetics , Green Fluorescent Proteins/genetics , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Animals , CHO Cells , Colforsin/pharmacology , Cricetinae , Cricetulus , Dinoprost/pharmacology , Female , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Haplorhini , Male , Mice, Inbred C57BL , Mice, Transgenic , Prolactin/pharmacology , Recombinant Fusion Proteins/metabolism
3.
Reprod Fertil Dev ; 29(11): 2175-2182, 2017 Oct.
Article En | MEDLINE | ID: mdl-28442026

The aims of the present study were to determine: (1) whether oestradiol (E2) in the superior mesenteric ganglion (SMG) modifies the release of ovarian progesterone (P4), androstenedione (A2) and E2, the activity and gene expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 20α-HSD and the expression of P450 aromatase (Cyp19a1) and (2) whether any such modifications are related to changes in ovarian nitric oxide (NO) and noradrenaline (NA) levels during dioestrus II. Using an ex vivo SMG-ovarian nervous plexus-ovary system, ovarian P4 release was measured following the addition E2 plus tamoxifen (Txf) (10-6M) to the ganglion, whereas A2, E2, NA and NO were measured following the addition of E2 alone. Steroids were measured by radioimmunoassay, NA concentrations were determined by HPLC and gene expression was evaluated using reverse transcription-polymerase chain reaction. Oestradiol in the ganglion decreased ovarian P4, E2 and NA release, as well as 3ß-HSD activity, but increased the release of A2 and nitrites, as well as the 20α-HSD expression and its activity. No changes were observed in Cyp19a1 gene expression. The addition of E2 plus Txf to the ganglion reversed the effects of E2 alone. The action of oestradiol in SMG favours the beginning of functional luteolysis, due to an increase in NO release and a decrease in NA in the ovary. These results may help elucidate the role of E2 in hormone-dependent pathologies in women.


Diestrus/drug effects , Estradiol/pharmacology , Ganglia, Sympathetic/drug effects , Nitric Oxide/metabolism , Ovary/drug effects , Progesterone/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Androstenedione/metabolism , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Diestrus/metabolism , Female , Ganglia, Sympathetic/metabolism , Norepinephrine/metabolism , Ovary/metabolism , Rats , Rats, Sprague-Dawley
4.
Reprod Fertil Dev ; 28(5): 565-73, 2016 Apr.
Article En | MEDLINE | ID: mdl-25194502

There is considerable evidence of the neuroendocrine control involved in luteal regression in the rat. In addition, circulating prolactin (PRL), which increases during the night before parturition, may gain access to the coeliac ganglion (CG), indirectly impacting the physiology of the ovary because of the known connection between the CG and the ovary via the superior ovarian nerve (SON). In this work we investigated in the CG-SON-ovary system and whether PRL added to the CG has an impact, indirectly via the SON, on luteal regression on Day 21 of pregnancy. The system was incubated without (control) or with PRL added to the CG. We measured the ovarian release of progesterone (P), oestradiol and prostaglandin F2 alpha (PGF2α) by radioimmunoassay, and nitrites (NO) by the Griess method. Luteal mRNA expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD), 20α-HSD, aromatase, inducible nitric oxide synthase (iNOS) and apoptosis regulatory factors was analysed by reverse transcription-polymerase chain reaction. P release, the expression of Bcl-2 and the Bcl-2:Bax ratio was lower than control preparations, while the expression of 20α-HSD and the release of NO and PGF2α were higher in the experimental group. In conclusion, PRL acts at the CG and, by a neural pathway, modulates luteal function at the end of pregnancy.


Corpus Luteum/innervation , Ganglia, Sympathetic/drug effects , Luteolysis/drug effects , Ovary/innervation , Prolactin/pharmacology , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Corpus Luteum/enzymology , Corpus Luteum/pathology , Dinoprost/metabolism , Estradiol/metabolism , Female , Ganglia, Sympathetic/physiology , Gestational Age , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Ovary/metabolism , Pregnancy , Progesterone/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Time Factors , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
J Steroid Biochem Mol Biol ; 154: 39-46, 2015 Nov.
Article En | MEDLINE | ID: mdl-26144997

The aim of this work was to investigate if noradrenaline (NA), added in the coeliac ganglion -superior ovarian nerve- ovary system (CG-SON-O) and in ovary incubation, modifies the release of ovarian progesterone (P4), gonadotropin-releasing hormone (GnRH) and oestradiol (E2), and the expression of 3ß-HSD and 20α-HSD and proapoptotic bax and antiapoptotic bcl-2 on dioestrus II in the rat. The CG-SON-O system and the ovary were removed and placed in one cuvette containing Krebs-Ringer solution (control groups), and NA was added to the ganglion compartment in the ex vivo system and in the ovary compartment in the ovary incubation (experimental groups). P4, GnRH and E2 were measured by RIA, and gene expression was measured by RT-PCR. In the ex-vivo system, the release of ovarian P4 and GnRH and the expression of 3ß-HSD and bax decreased; E2 and bcl-2 increased, and the bax/bcl-2 ratio decreased. However, in the ovary incubation, P4, GnRH, the expression of 3ß-HSD and bax increased; E2, the expression of 20α-HSD and bcl-2 decreased while the bax/bcl-2 ratio increased, thus favoring apoptosis. The peripheral nervous system protected the ovary from the apoptotic mechanisms while in the ovary incubation the effect was reverted. Our results indicate that NA regulates ovarian steroidogenesis and apoptosis by modulating GnRH release from the coeliac ganglion and ovary, being NA a possible generator of a GnRH-gonadotropins axis in the ovary. This work is expected to contribute with new evidence of the clinical importance of catecholamines and GnRH in therapy and prevention of ovarian pathologies.


Apoptosis/drug effects , Gonadotropin-Releasing Hormone/metabolism , Norepinephrine/pharmacology , Ovary/drug effects , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Estradiol/biosynthesis , Female , Gonadotropin-Releasing Hormone/biosynthesis , Ovary/enzymology , Ovary/metabolism , Progesterone/biosynthesis , Rats , Rats, Sprague-Dawley
6.
Anim Reprod Sci ; 148(1-2): 63-71, 2014 Jul.
Article En | MEDLINE | ID: mdl-24875897

The aims of this study were to determine the specific site of 20α-HSD expression in the reproductive tissues on day 30 of pregnancy and during pre-parturition. 20α-HSD mRNA was demonstrated to have the highest expression in the placenta on day 30 of pregnancy and in the ovary during pre-parturition. Weak mRNA expression was observed in the uterus and ovary on day 30 of pregnancy. However, the mRNA was not expressed in the oviduct on day 30 of pregnancy. The mRNA was also specifically detected in the placenta on day 30 of pregnancy by northern blot analysis. Western blot analysis indicated that the expression pattern of the 20α-HSD protein in the reproductive tissues was similar to that of 20α-HSD mRNA. Immunohistochemical analysis also revealed that the pig 20α-HSD protein was localized in the trophoblast villus in the placenta on day 30 of pregnancy. It was highly expressed in the glandular epithelial cells of the endometrium and the luminal epithelial cells of the uterus. The 20α-HSD protein was highly localized in the large luteal cells of the ovary on day 30 of pregnancy and during pre-parturition. Taken together, our study demonstrated that the pig 20α-HSD mRNA and protein are mainly localized in the trophoblast villus in the placenta on day 30 of pregnancy. The expression of the protein is also localized in the large luteal cells of the ovary. In addition, the protein is highly expressed in the glandular epithelial cells of the endometrium and the luminal epithelial cells of the uterus.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Gene Expression Regulation, Enzymologic/physiology , Pregnancy, Animal , Swine/physiology , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Animals , Female , Ovary/metabolism , Placenta/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Distribution
7.
BMC Res Notes ; 7: 119, 2014 Mar 01.
Article En | MEDLINE | ID: mdl-24580729

BACKGROUND: Glucocorticoids exert recognized positive effects on lung development. The genes involved in the classical pathway of glucocorticoid synthesis normally occurring in adrenals were found to be expressed on gestation day (GD) 15.5 in the developing mouse lung. Recently, expression of two of these genes was also detected on GD 17.5 suggesting a more complex temporal regulation than previously expected. Here, we deepen the knowledge on expression of "adrenal" glucocorticoid synthesis genes in the mouse lung during the perinatal period and we also study expression of the gene encoding for the steroid inactivating enzyme 20α-hydroxysteroid dehydrogenase (20α-HSD). RESULTS: We performed an ontogenic study of P450scc, 3ß-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase 1 (3ß-HSD1), 21-hydroxylase, 11ß-hydroxylase, 11ß-HSD1, and 11ß-HSD2 expression up to post natal day (PN) 15. The substrate (progesterone) and the product (deoxycorticosterone) of 21-hydroxylase are substrates of 20α-HSD, thus 20α-HSD (Akr1c18) gene expression was investigated. In lung samples collected between GD 15.5 and PN 15, 11ß-hydroxylase was only detected on GD 15.5. In contrast, all the other tested genes were expressed throughout the analyzed period with different temporal expression patterns. P450scc, 21-hydroxylase, 20α-HSD and 11ß-HSD2 mRNA levels increased after birth with different patterns including an increase from PN 3 with a possible sex difference for 21-hydroxylase mRNA. Also, the 21-hydroxylase protein was observed by Western blot in perinatal lungs with higher levels after birth. CONCLUSION: Progesterone is present at high levels during gestation and the product of 21-hydroxylase, deoxycorticosterone, can bind the glucocorticoid receptor with an affinity close to that of corticosterone. Detection of 21-hydroxylase at the protein level during antenatal lung development is the first evidence that the adrenal-like glucocorticoid synthesis pathway detected during lung development has the machinery to produce glucocorticoids in the fetal lung. Glucocorticoids from lung 21-hydroxylase appear to modulate lung ontogenesis through paracrine/intracrine actions.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Adrenal Cortex Hormones/biosynthesis , Glucocorticoids/biosynthesis , Lung/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Animals , Biosynthetic Pathways/genetics , Blotting, Western , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Lung/embryology , Lung/growth & development , Male , Mice, Inbred BALB C , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/metabolism
8.
Biol Pharm Bull ; 36(9): 1514-8, 2013.
Article En | MEDLINE | ID: mdl-23995665

In this study, we examined the substrate specificity and inhibitor sensitivity of rabbit 20α-hydroxysteroid dehydrogenase (AKR1C5), which plays a role in the termination of pregnancy by progesterone inactivation. AKR1C5 moderately reduced the 3-keto group of only 5α-dihydrosteroids with 17ß- or 20α/ß-hydroxy group among 3-ketosteroids. In contrast, the enzyme reversibly and efficiently catalyzed the reduction of various 17- and 20-ketosteroids, including estrogen precursors (dehydroepiandrosterone, estrone and 5α-androstan-3ß-ol-17-one) and tocolytic 5ß-pregnane-3,20-dione. In addition to the progesterone inactivation, the formation of estrogens and metabolism of the tocolytic steroid by AKR1C5 may be related to its role in rabbit parturition. AKR1C5 also reduced various non-steroidal carbonyl compounds, including isatin, an antagonist of the C-type natriuretic peptide receptor, and 4-oxo-2-nonenal, suggesting its roles in controlling the bioactive isatin and detoxification of cytotoxic aldehydes. AKR1C5 was potently and competitively inhibited by flavonoids such as kaempferol and quercetin, suggesting that its activity is affected by ingested flavonoids.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/antagonists & inhibitors , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Aldehydes/metabolism , Animals , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hydroxysteroids/metabolism , Isatin/metabolism , Ketones/metabolism , Ketosteroids/metabolism , Quinones/metabolism , Rabbits , Recombinant Proteins/metabolism , Substrate Specificity , Xenobiotics/metabolism
9.
Fertil Steril ; 99(7): 2062-70, 2013 Jun.
Article En | MEDLINE | ID: mdl-23517861

OBJECTIVE: To investigate whether cholinergic ganglionic stimulus modifies the release of gonadotropin-releasing hormone (GnRH), catecholamines, and progesterone at the ovarian level. DESIGN: Animal study. SETTING: University animal laboratory. ANIMAL(S): Six to eight virgin adult Holtzman rats. INTERVENTION(S): Superior mesenteric ganglion-ovarian nerve plexus-ovary system removed and placed in one cuvette with two compartments, with acetylcholine added to the ganglion in the experimental group. MAIN OUTCOME MEASURE(S): Measurement of ovarian liquid obtained from catecholamines by high-performance liquid chromatography; measurement of progesterone (P(4)), GnRH, and luteinizing hormone (LH) by radioimmunoassay; and measurement of gene expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 20α-hydroxysteroid dehydrogenase (20α-HSD) by reverse-transcriptase polymerase chain reaction (RT-PCR). RESULT(S): The study focused on the estrus and diestrus II (DII) stages. On the estrus days, the release of GnRH, NA, and 20α-HSD increased, while P(4) and 3ß-HSD decreased. On the DII days, GnRH, P(4), and 3ß-HSD increased, while 20α-HSD and NA decreased. The ovarian liquid with GnRH showed biologic activity, namely, an increase in LH release during the DII stage and a decrease during the estrus stage. CONCLUSION(S): Neural stimulus from the superior mesenteric ganglion influences the release of NA, adrenaline, and GnRH. We also have demonstrated that these neurotransmitters participate in the atretogenic processes of the ovary, thus providing evidence of the necessity of the sympathetic neural pathway.


Catecholamines/metabolism , Ganglia, Sympathetic/metabolism , Gonadotropin-Releasing Hormone/metabolism , Ovary/innervation , Ovary/metabolism , Progesterone/metabolism , Receptors, Cholinergic/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Acetylcholine/metabolism , Animals , Chromatography, High Pressure Liquid , Diestrus/metabolism , Estrus/metabolism , Female , Ovary/enzymology , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
10.
Gen Comp Endocrinol ; 184: 1-8, 2013 Apr 01.
Article En | MEDLINE | ID: mdl-23313075

Whether prolactin (PRL) has a luteotrophic or luteolytic effect in the rat ovary depends on the nature of the corpora lutea present in the ovaries and the hormonal environment to which they are exposed. The aim was to investigate the effect of PRL acting on the coeliac ganglion (CG) on the function of the corpora lutea on day 4 postpartum under either lactating or non-lactating conditions, using the CG-superior ovarian nerve-ovary system. The ovarian release of progesterone (P), estradiol, PGF2α, and nitrites was assessed in the ovarian compartment at different incubation times. Luteal mRNA expression of 3ß-HSD, 20α-HSD, aromatase, PGF2α receptor, iNOS, Bcl-2, Bax, Fas and FasL was analysed in the corpus luteum of pregnancy at the end of the experiments. Comparative analysis of control groups showed that the ovarian release of P, nitrites, and PGF2α, the expression of PGF2α receptor, and the Bcl-2/Bax ratio were lower in non-lactating rats, with increased release of estradiol, and higher expression of aromatase, Fas and FasL, demonstrating the higher luteal functionality in ovaries of lactating animals. PRL added to the CG compartment increased the ovarian release of P, estradiol, nitrites and PGF2α, and decreased the Bcl-2/Bax ratio in non-lactating rats; yet, with the exception of a reduction in the release of nitrites, such parameters were not modified in lactating animals. Together, these data suggest that the CG is able to respond to the effect of PRL and, via a neural pathway, fine-tune the physiology of the ovary under different hormonal conditions.


Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Lactation/drug effects , Lactation/metabolism , Ovary/innervation , Ovary/metabolism , Postpartum Period/metabolism , Prolactin/pharmacology , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Aromatase/genetics , Estradiol/metabolism , Fas Ligand Protein/genetics , Female , Nitrites/metabolism , Ovary/drug effects , Postpartum Period/drug effects , Pregnancy , Progesterone/metabolism , Prostaglandins/metabolism , Radioimmunoassay , Rats , Receptors, Prostaglandin/genetics , Reverse Transcriptase Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics
11.
J Steroid Biochem Mol Biol ; 135: 60-6, 2013 May.
Article En | MEDLINE | ID: mdl-23313240

The aims of this work were to investigate if oestradiol 10(-8)M in the incubation media of either the ovary alone (OV) or the ganglion compartment of an ex vivo coeliac ganglion-superior ovarian nerve-ovary system (a) modifies the release of ovarian progesterone (P4) and oestradiol (E2) on dioestrus II, and (b) modifies the ovarian gene expression of 3ß-HSD and 20α-HSD enzymes and markers of apoptosis. The concentration of ovarian P4 release was measured in both experimental schemes, and ovarian P4 and E2 in the ex vivo system by RIA at different times. The expression of 3ß-hydroxysteroid dehydrogenase, 20α-hydroxysteroid dehydrogenase and antiapoptotic bcl-2 and proapoptotic bax by RT-PCR were determined. E2 added in the coeliac ganglion caused an increase in the ovarian release of the P4, E2 and 3ß-HSD, while in the ovary incubation alone it decreased P4 and 3ß-HSD but increased and 20α-HSD and bax/bcl-2 ratio. It is concluded that through a direct effect on the ovary, E2 promotes luteal regression in DII rats, but the addition of E2 in the coeliac ganglion does not have the same effect. The peripheral nervous system, through the superior ovarian nerve, has a protective effect against the apoptotic mechanism on DII.


Apoptosis/drug effects , Diestrus , Estradiol/pharmacology , Ganglia, Sympathetic/drug effects , Ovary , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Estradiol/biosynthesis , Estradiol/metabolism , Female , Ganglia, Sympathetic/metabolism , Ovary/drug effects , Ovary/innervation , Ovary/metabolism , Progesterone/metabolism , Rats , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/metabolism
12.
Proc Natl Acad Sci U S A ; 109(19): 7529-34, 2012 May 08.
Article En | MEDLINE | ID: mdl-22529366

During pregnancy, uterine quiescence is maintained by increased progesterone receptor (PR) activity, but labor is facilitated by a series of events that impair PR function. Previously, we discovered that miR-200 family members serve as progesterone (P(4))-modulated activators of contraction-associated genes in the pregnant uterus. In this study, we identified a unique role for miR-200a to enhance the local metabolism of P(4) in myometrium and, thus, decrease PR function during the progression toward labor. miR-200a exerts this action by direct repression of STAT5b, a transcriptional repressor of the P(4)-metabolizing enzyme 20α-hydroxysteroid dehydrogenase (20α-HSD). We observed that miR-200a expression increased and STAT5b expression coordinately decreased in myometrium of mice as they progressed to labor and in laboring myometrium from pregnant women. These changes were associated with a dramatic increase in expression and activity of 20α-HSD in laboring myometrium from mouse and human. Notably, overexpression of miR-200a in cultured human myometrial cells (hTERT-HM) suppressed STAT5b and increased 20α-HSD mRNA levels. In uterine tissues of ovariectomized mice injected with P(4), miR-200 expression was significantly decreased, STAT5b expression was up-regulated, and 20α-HSD mRNA was decreased, but in 15 d postcoitum pregnant mice injected with the PR antagonist RU486, preterm labor was associated with increased miR-200a, decreased STAT5b, and enhanced 20α-HSD expression. Taken together, these findings implicate miR-200a as an important regulator of increased local P(4) metabolism in the pregnant uterus near term and provide insight into the importance of miR-200s in the decline in PR function leading to labor.


Labor, Obstetric/genetics , MicroRNAs/genetics , Obstetric Labor, Premature/genetics , Receptors, Progesterone/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Animals , Cells, Cultured , Female , Gene Expression/drug effects , Hormone Antagonists/pharmacology , Humans , Immunoblotting , Labor, Obstetric/metabolism , Male , Mice , Mice, Inbred ICR , Mifepristone/pharmacology , Myometrium/cytology , Myometrium/metabolism , Obstetric Labor, Premature/metabolism , Ovariectomy , Pregnancy , Progesterone/metabolism , Progesterone/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Uterus/drug effects , Uterus/metabolism
13.
Reprod Sci ; 19(4): 416-22, 2012 Apr.
Article En | MEDLINE | ID: mdl-22101240

There is evidence suggesting that estradiol (E(2)) regulates the physiology of the ovary and the sympathetic neurons associated with the reproductive function. The objective of this study was to investigate the effect of E(2) on the function of late pregnant rat ovaries, acting either directly on the ovarian tissue or indirectly via the superior ovarian nerve (SON) from the celiac ganglion (CG). We used in vitro ovary (OV) or ex vivo CG-SON-OV incubation systems from day 21 pregnant rats. Various concentrations of E(2 )were added to the incubation media of either the OV alone or the ganglion compartment of the CG-SON-OV system. In both experimental schemes, we measured the concentration of progesterone in the OV incubation media by radioimmunoassay at different times. Luteal messenger RNA (mRNA) expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 20α-hydroxysteroid dehydrogenase (20α-HSD) enzymes, respectively, involved in progesterone synthesis and catabolism, and of antiapoptotic B-cell lymphoma 2 (Bcl-2) and proapoptotic Bcl-2-associated X protein (Bax), were measured by reverse transcriptase-polymerase chain reaction (RT-PCR) at the end of the incubation period. Estradiol added directly to the OV incubation or to the CG of the CG-SON-OV system caused a decline in the concentration of progesterone accumulated in the incubation media. In addition, E(2), when added to the OV incubation, decreased the expression of 3ß-HSD and the ratio of Bcl-2/Bax. We conclude that through a direct effect on the OV, E(2) favors luteal regression at the end of pregnancy in rats, in association with neural modulation from the CG via the SON.


Corpus Luteum/drug effects , Estradiol/pharmacology , Ganglia, Sympathetic/drug effects , Luteolysis/drug effects , Ovary/drug effects , Progesterone/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Corpus Luteum/enzymology , Corpus Luteum/innervation , Corpus Luteum/physiology , Female , Ganglia, Sympathetic/enzymology , Ganglia, Sympathetic/physiology , In Vitro Techniques , Luteolysis/physiology , Ovary/enzymology , Ovary/innervation , Ovary/physiology , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
14.
Reprod Biol Endocrinol ; 9: 139, 2011 Oct 20.
Article En | MEDLINE | ID: mdl-22014308

BACKGROUND: The aldo-keto reductase family 1 member C1 (AKR1C1) belongs to a superfamily of NADPH-dependent reductases that convert a wide range of substrates, including carbohydrates, steroid hormones, and endogenous prostaglandins. The 20 alpha-hydroxysteroid dehydrogenase (20 alpha-HSD) is a member of AKR family. The aims of this study were to determine its expression in the ovary and uterus endometrium during the estrous cycle and pregnancy. METHODS: Rapid amplification of cDNA ends (RACE) experiments were performed to obtain the 5' and 3' ends of the porcine 20 alpha-HSD cDNA. Reverse-transcriptase-PCR (RT-PCR), real-time PCR, northern blot analysis, and western blot analysis were performed to examine the expression of porcine 20 alpha-HSD. Immunohistochemical analysis was also performed to determine the localization in the ovary. RESULTS: The porcine 20 alpha-HSD cDNA is 957 bp in length and encodes a protein of 319 amino acids. The cloned cDNA was virtually the same as the porcine AKR1C1 gene (337 amino acids) reported recently, and only differed in the C-terminal region (the AKR1C1 gene has a longer C-terminal region than our sequence). The 20 alpha-HSD gene (from now on referred to as AKR1C1) cloned in this paper encodes a deletion of 4 amino acids, compared with the C-terminal region of AKR1C1 genes from other animals. Porcine AKR1C1 mRNA was expressed on day 5, 10, 12, 15 of the cycle and 0-60 of pregnancy in the ovary. The mRNA was also specifically detected in the uterine endometrium on day 30 of pregnancy. Western blot analysis indicated that the pattern of AKR1C1 protein in the ovary during the estrous cycle and uterus during early pregnancy was similar to that of AKR1C1 mRNA expression. The recombinant protein produced in CHO cells was detected at approximately 37 kDa. Immunohistochemical analysis also revealed that pig AKR1C1 protein was localized in the large luteal cells in the early stages of the estrous cycle and before parturition. CONCLUSIONS: Our study demonstrated that AKR1C1 mRNA and protein are coordinately expressed in the luteal cell of ovary throughout the estrous cycle and in the uterus on day 30 of pregnancy. Thus, the porcine AKR1C1 gene might control important mechanisms during the estrous cycle.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Endometrium/metabolism , Estrous Cycle/metabolism , Ovary/metabolism , Pregnancy Proteins/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/chemistry , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Amino Acid Sequence , Animals , Cell Size , Codon, Terminator , Databases, Nucleic Acid , Female , Gene Expression Regulation, Enzymologic , Luteal Cells/metabolism , Molecular Sequence Data , Molecular Weight , Ovary/cytology , Pregnancy , Pregnancy Proteins/chemistry , Pregnancy Proteins/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Sus scrofa
15.
Reproduction ; 142(5): 723-31, 2011 Nov.
Article En | MEDLINE | ID: mdl-21908655

The enzyme 20α-hydroxysteroid dehydrogenase (20α-HSD) catalyzes the conversion of progesterone to its inactive form, 20α-hydroxyprogesterone. This enzyme plays a critical role in the regulation of luteal function in female mammals. In this study, we conducted the characterization and functional analyses of bovine 20α-HSD from placental and ovarian tissues. The nucleotide sequence of bovine 20α-HSD showed significant homology to that of goats (96%), humans (84%), rabbits (83%), and mice (81%). The mRNA levels increased gradually throughout the estrous cycle, the highest being in the corpus luteum (CL) 1 stage. Northern blot analysis revealed a 1.2  kb mRNA in the bovine placental and ovarian tissues. An antibody specific to bovine 20α-HSD was generated in a rabbit immunized with the purified, recombinant protein. Recombinant 20α-HSD protein produced in mammalian cells had a molecular weight of ∼37  kDa. Bacterially expressed bovine 20α-HSD protein showed enzymatic activity. The expression pattern of the 20α-HSD protein in the pre-parturition placenta and the CL1 stage of the estrous cycle was similar to the level of 20α-HSD mRNA expression. Immunohistochemical analysis also revealed that bovine 20α-HSD protein was intensively localized in the large luteal cells during the late estrous cycle.


20-alpha-Hydroxysteroid Dehydrogenase/isolation & purification , Ovary/enzymology , Placenta/enzymology , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cattle , Cloning, Molecular , Cricetinae , Cricetulus , Female , Gene Expression Regulation, Enzymologic , Mice , Molecular Sequence Data , Ovary/chemistry , Ovary/metabolism , Phylogeny , Placenta/chemistry , Placenta/metabolism , Pregnancy , Rabbits , Rats , Sequence Analysis, DNA , Sequence Homology, Amino Acid
16.
J Steroid Biochem Mol Biol ; 127(3-5): 337-44, 2011 Nov.
Article En | MEDLINE | ID: mdl-21831350

This study was conducted to characterize and functionally analyze the monkey 20α-hydroxysteroid dehydrogenase (20α-HSD) in the ovary, placenta, and oviduct. We focused on 20α-HSD mRNA expression and protein localization in monkey reproductive tissues and the molecular characterization of the promoter region. Reverse transcription-polymerase chain reaction (RT-PCR) monkey 20α-HSD mRNA was more strongly detected in the ovary at pre-ovulation than in the placenta and oviduct at pre-parturition. The mRNA was approximately 1.2kb in size and the expression was high in the ovary, which was the same as the RT-PCR result. We also produced His tagged 20α-HSD proteins by using an Escherichia coli expression system. In a western blot for the 20α-HSD protein, only 1 band of approximately 37-kDa was detected in the ovary, oviduct tissue, and recombinant protein produced in the Chinese hamster ovary (CHO) cell line. However, in the placenta, additional 2 bands (35 and 39 kDa) were detected. Immunohistochemical analyses suggested that the monkey 20α-HSD protein was localized mainly in the syncytiotrophoblast of the placenta and the isthmus cells of the oviduct. According to promoter analyses with the enhanced green fluorescent protein (EGFP) gene, the monkey 20α-HSD promoter was efficiently expressed in the CHO-K1 cell line; however, the promoter was not expressed in bovine fetal fibroblast (bFF) cell. Taken together, our study showed that the 20α-HSD mRNA and protein are coordinately expressed in the ovary at pre-ovulation and in the placenta and oviduct at pre-parturition. Therefore, monkey 20α-HSD in the placenta, ovary and oviduct plays an important role in the estrous cycle, pregnancy, and parturition.


20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Ovary/enzymology , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Animals , Base Sequence , Blotting, Northern , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , DNA Primers , Female , Immunohistochemistry , Macaca fascicularis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Appl Biochem Biotechnol ; 165(1): 190-203, 2011 Sep.
Article En | MEDLINE | ID: mdl-21494755

The human sex hormone progesterone plays an essential and complex role in a number of physiological processes. Progesterone deficiency is associated with menstrual disorders and infertility as well as premature birth and abortion. For progesterone replacement therapy, the synthetic progestogen dydrogesterone is commonly used. In the body, this drug is metabolized to 20α-dihydrodydrogesterone (20α-DHD), which also shows extensive pharmacological effects and hence could act as a therapeutic agent itself. In this study, we describe an efficient biotechnological production procedure for 20α-DHD that employs the stereo- and regioselective reduction of dydrogesterone in a whole-cell biotransformation process based on recombinant fission yeast cells expressing the human enzyme AKR1C1 (20α-hydroxysteroid dehydrogenase, 20α-HSD). In a fed-batch fermentation at pilot scale (70 L) with a genetically improved production strain and under optimized reaction conditions, an average 20α-DHD production rate of 190 µM day(-1) was determined for a total biotransformation time of 136 h. Combined with an effective and reliable downstream processing, a continuous production rate of 12.3 ± 1.4 g 20α-DHD per week and fermenter was achieved. We thus established an AKR-dependent whole-cell biotransformation process that can also be used for the production of other AKR1C1 substrates (as exemplarily shown by the production of 20α-dihydroprogesterone in gram scale) and is in principle suited for the production of further human AKR metabolites at industrial scale.


Biotechnology/methods , Dydrogesterone/analogs & derivatives , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Dydrogesterone/metabolism , Fermentation/physiology , Humans , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
18.
J Anim Sci ; 89(5): 1338-46, 2011 May.
Article En | MEDLINE | ID: mdl-21521814

In the present study, we performed quantitative reverse-transcription PCR (qPCR) to examine changes in gene expression of prolactin receptor (long form: l-PRLR; short form: s-PRLR) and 20α-hydroxysteroid dehydrogenase (20α-HSD; EC 1.1.1.149) in the bovine corpus luteum (CL) throughout the estrous cycle and pregnancy. Western blotting was used to determine protein abundance. Bovine CL were collected and luteal stages (n = 6/stage) were classified by macroscopic observation as early (d 1 to 4 after ovulation), mid (d 5 to 10), late (d 11 to 17), and regressing (d 18 to 20). A CL of pregnancy (n = 6) was determined by the presence of conceptus (d 28 to term). The mRNA for both forms of PRLR were expressed at all the luteal stages. Expression of s-PRLR and l-PRLR mRNA was less (P < 0.01) during early and regressing luteal stages compared with mid and late stages. Expression of s-PRLR mRNA in CL of pregnancy was greater (P < 0.01) than early, mid, and regressing CL and did not differ from late luteal stage expression. A greater (P < 0.01) expression of l-PRLR mRNA was observed in pregnant vs. early and regressing CL. In addition, qPCR showed the presence of 20α-HSD mRNA during all luteal stages of the estrous cycle, with the greatest (P < 0.01) expression observed in the regressing luteal stage. Western blotting revealed protein abundance of both PRLR isoforms during all luteal stages and pregnancy, with a predominance of the s-PRLR protein. Densitometry analysis indicated that protein abundances of s-PRLR were greater (P < 0.05) than l-PRLR during early, mid, and late luteal stages and did not differ during the regressing luteal stage. Protein abundances of 20α-HSD were least (P < 0.05) during the early luteal stage. In conclusion, results of the current study suggest a possible involvement of PRLR, especially s-PRLR, in the regulation of progesterone secretion and metabolism during the bovine estrous cycle and pregnancy.


Cattle/physiology , Corpus Luteum/metabolism , Estrous Cycle/physiology , Progesterone/metabolism , Receptors, Prolactin/biosynthesis , 20-alpha-Hydroxysteroid Dehydrogenase/biosynthesis , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Animals , Blotting, Western/veterinary , Corpus Luteum/enzymology , Female , Gene Expression Regulation, Enzymologic , Pregnancy , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Prolactin/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
19.
Toxicol Sci ; 121(2): 267-78, 2011 Jun.
Article En | MEDLINE | ID: mdl-21427058

Ethylene glycol monomethyl ether (EGME), sulpiride, and atrazine are known ovarian toxicants, which increase progesterone (P4) secretion and induce luteal cell hypertrophy following repeated administration. The aim of this study was to define the pathways by which these compounds exerted their effects on the ovary and hypothalamic-pituitary-gonadal (HPG) axis. In the ovary, changes in the steroidogenic activity of new and old corpora lutea (CL) were addressed. EGME (300 mg/kg), sulpiride (100 mg/kg), or atrazine (300 mg/kg) were orally given daily for four times from proestrus to diestrus in normal cycling rats. Treatment with all chemicals significantly increased serum P4 levels, and EGME as well as sulpiride induced increases in prolactin (PRL) levels. In new CL, at both the gene and the protein levels, all three chemicals upregulated the following steroidogenic factors: scavenger receptor class B type I, steroidogenic acute regulatory protein, P450 cholesterol side-chain cleavage, and 3ß-hydroxysteroid dehydrogenase (HSD) and downregulated the luteolytic gene, 20α-HSD. Coadministration of EGME and bromocriptine, a D2 agonist, completely inhibited PRL but not P4 secretion. Additionally, steroidogenic factor expression levels were upregulated, and 20α-HSD level was downregulated in new CL. These results suggest that EGME both directly and indirectly stimulates P4 production in luteal cells, whereas sulpiride elevates P4 through activation of PRL secretion in the pituitary. Atrazine may directly activate new CL by stimulating steroidogenic factor expressions. The present study suggests that multiple pathways mediate the effects of EGME, sulpiride, and atrazine on the HPG axis and luteal P4 production in female rats in vivo.


Atrazine/toxicity , Ethylene Glycols/toxicity , Luteal Cells/drug effects , Progesterone/metabolism , Sulpiride/toxicity , 17-Hydroxysteroid Dehydrogenases/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Amino Acid Sequence , Animals , Bromocriptine/toxicity , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Down-Regulation , Female , Immunohistochemistry , Microdissection/methods , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Progesterone/blood , Prolactin/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
20.
J Steroid Biochem Mol Biol ; 124(1-2): 58-64, 2011 Mar.
Article En | MEDLINE | ID: mdl-21262361

The objective of the present study was to examine some factors involved in follicular development of women with polycystic ovary syndrome (PCOS). Women with PCOS showed increased levels of serum luteinizing hormone (LH) but decreased follicular production of progesterone and estradiol by pre-ovulatory follicles. The mRNA expression corresponding to steroidogenic acute regulatory protein (StAR), and 20alpha-hydroxysteroid dehydrogenase (20α-HSD) was increased, while that corresponding to cytochrome P450 aromatase (P450arom) was decreased in PCOS follicles as compared to controls. No changes in the mRNA expression for 3beta-hydroxysteroid dehydrogenase 2 (3ß-HSD2), cytochrome P450 side-chain cleavage (P450scc), cytochrome P450 17 alpha hydroxylase/lyase (P450c17), cyclooxygenase 2 (COX2), and transcription factors (GATA-4 and GATA-6) were found. We conclude that despite the hyper-luteinized environment of PCOS follicles, these follicles produce lower levels of progesterone and estradiol, and that this is characterized by increased degradation of progesterone and decreased estradiol synthesis. Our data demonstrate that the synthesis of prostaglandin F2α (PGF2α) may be affected in PCOS-follicles and that the transcription factors GATA-4 and GATA-6 are present in PCOS-follicles but they are not involved in the abnormal transcription observed in the steroidogenic enzymes.


Ovarian Follicle/pathology , Polycystic Ovary Syndrome/pathology , 20-alpha-Hydroxysteroid Dehydrogenase/biosynthesis , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 3-Hydroxysteroid Dehydrogenases/biosynthesis , 3-Hydroxysteroid Dehydrogenases/genetics , Aromatase/biosynthesis , Aromatase/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Estradiol/biosynthesis , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/physiology , Humans , Luteinizing Hormone/blood , Ovarian Follicle/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/metabolism , Progesterone/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroid 17-alpha-Hydroxylase/biosynthesis , Steroid 17-alpha-Hydroxylase/genetics
...