Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Phytother Res ; 35(11): 6170-6180, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33908658

ABSTRACT

It is widely known that breast cancer cells eventually develop resistance to hormonal drugs and chemotherapies, which often compromise fertility. This study aimed to investigate the effect of the flavonoid, kaempferol-3-O-apiofuranosyl-7-O-rhamnopyranosyl (KARP), on 1) the viability of MCF-7 breast cancer cells and 2) ovarian function in rats. A dose-dependent decrease in MCF-7 cell survival was observed, and the IC50 value was found to be 48 µg/ml. Cells in the control group or those exposed to increasing concentrations of KARP experienced a similar generation of reactive oxygen species and induction of apoptosis. For the rats, estradiol levels correlated negatively to KARP dosages, although a recovery was obtained at administration of 30 mg/kg per day. Noteworthily, when compared against the control, this dosage led to significant increases in mRNA levels for CYP19, CYP17a, CCND2, GDF9, and INSL3 among the treatment groups, and ER1 and ER2 mRNA levels decreased in a dose-dependent manner. KARP shows great promise as an ideal therapy for breast cancer patients since it induced apoptosis and autophagy in cancerous cells without harming fertility in our animal model. Future investigations on humans are necessary to substantiate these findings and determine its efficacy as a general line of treatment.


Subject(s)
Breast Neoplasms , Flavonoids , Animals , Apoptosis , Aromatase/genetics , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cyclin D2 , Female , Growth Differentiation Factor 9/genetics , Humans , Insulin/genetics , Kaempferols/pharmacology , Proteins/genetics , Rats , Steroid 17-alpha-Hydroxylase/genetics
2.
J Int Med Res ; 49(3): 300060521993965, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33761789

ABSTRACT

BACKGROUND: We report here a case study of 17α-hydroxylase deficiency in a phenotypic girl with male karyotype (46,XY). We also review the relevant literature to deepen our understanding of the disease, reduce the rate of missed diagnosis, and emphasize that holistic management of this disease requires collaborative multidisciplinary teamwork. CASE PRESENTATION: A 14-year-old patient with a female phenotype visited the endocrinology department because of hypertension. The patient had primary amenorrhea and lacked secondary sexual characteristics. Initial laboratory evaluation revealed normal levels of electrolytes, a hypergonadotropic hypogonadal state with high progesterone and low testosterone levels, and a 46,XY karyotype. She was referred to the urology department for gonadectomy and transferred to the gynecological endocrine clinic. On the basis of the patient's medical history and genetic testing results, a diagnosis of 46,XY 17α-hydroxylase deficiency was made. The patient was provided with glucocorticoids, estrogens, metformin, and psychological support. CONCLUSIONS: Patients with 17α-hydroxylase deficiency, a rare cause of congenital adrenal hyperplasia, should be treated by a multidisciplinary team. Relevant experts from different disciplines should set up a systematic and comprehensive individualized management plan to optimize the physical and mental health and quality of life of affected patients.


Subject(s)
Adrenal Hyperplasia, Congenital , Steroid 17-alpha-Hydroxylase , Adolescent , Adrenal Hyperplasia, Congenital/diagnosis , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/genetics , Female , Humans , Male , Mutation , Patient Care Team , Quality of Life , Steroid 17-alpha-Hydroxylase/genetics
3.
J Steroid Biochem Mol Biol ; 204: 105750, 2020 11.
Article in English | MEDLINE | ID: mdl-32920127

ABSTRACT

Ghrelin is a 28-amino acid peptide hormone that regulates ovarian steroid hormone synthesis; however, there is limited evidence regarding the regulation of this pathway by ghrelin in mice ovary. Thus, we aimed to investigate whether central ghrelin action plays a role in murine reproductive health by inhibiting steroid synthesis. Further, we sought to examine the mechanism of central ghrelin action in ovarian steroid hormone synthesis. After the administration of intracerebroventricular ghrelin (1 nmol), we found reduced serum concentrations of oestradiol and progesterone and reduced secretion of follicle-stimulating hormone and luteinising hormone. Although ghrelin reduced 3ß-hydroxysteroid dehydrogenase mRNA and protein levels in the hypothalamus, it did not affect the expression of steroidogenic acute regulatory protein and cytochrome P450 17A1. In the ovary, central ghrelin regulation indirectly inhibited the mRNA and protein levels of steroidogenic acute regulatory protein, cytochrome P450 17A1, and 3ß-hydroxysteroid dehydrogenase. Moreover, no changes were observed in the expression of proliferating cell nuclear antigen and phosphorylation of extracellular signal-regulated kinase. We hypothesised that central ghrelin regulation suppressed serum oestradiol and progesterone levels by indirectly inhibiting the expression of steroidogenic acute regulatory protein, cytochrome P450 17A1, and 3ß-hydroxysteroid dehydrogenase in the ovary. In this regulation, the suppressed secretion of the follicle-stimulating hormone and luteinising hormone in the pituitary by ghrelin could be involved. Furthermore, hypothalamic 3ß-hydroxysteroid dehydrogenase expression is reduced by ghrelin injection.


Subject(s)
Ghrelin/metabolism , Hormones/blood , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Female , Hypothalamus/metabolism , Injections, Intraventricular , Mice, Inbred C57BL , Ovary/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reproduction , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
4.
Environ Sci Pollut Res Int ; 26(23): 23306-23318, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31190303

ABSTRACT

The impact of Moringa oleifera leaf ethanol extract (MOLEE) was assessed on the expression of the steroidogenic genes (steroidogenic acute regulatory protein (StAR) and cytochrome P450c17 subfamily a (CYP17a) and luteinizing hormone receptor (LHR) gene) as well as on the cadmium chloride (CdCl2)-induced reproductive toxicity for 56 days in male rats. Four groups were used: control, Moringa-treated (MOLEE), CdCl2-treated, and CdCl2 + MOLEE groups. The reproductive toxicity of CdCl2 was confirmed; it caused a significant decrease in the accessory sex organ weights, testosterone level, testicular GST level, elevated MDA level (lipid peroxidation indicator), and histopathological alterations in seminiferous tubules, prostate, seminal vesicles, and epididymis as well as sperm characteristics. It also induced downregulation in the expression of StAR and CYP17a genes without change in the expression LHR gene. Eleven active compounds were detected in the GC-MS analysis of MOLEE; six of them have antioxidant properties, and five new compounds presented variable activities. MOLEE alone induced a stimulatory effect on the expression of steroidogenic and LHR genes. It restored the weight of reproductive organs to the control level; however, the recovery in sperm count, motility, abnormalities, percentage of alive sperm, testosterone, and MDA level are still comparable with the control level. Similar findings were also reported at the histological structure of the testes, epididymis, and accessory sex glands. Complete recovery of the GST enzyme activity was observed. Additionally, a restoration in the expression level of the steroidogenic genes was also reported. Our results indicated that the concurrent administration of MOLEE with CdCl2 can partially mitigate its harmful effects on male fertility.


Subject(s)
Cadmium Chloride/toxicity , Moringa/chemistry , Phosphoproteins/genetics , Plant Extracts/pharmacology , Reproduction/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Animals , Down-Regulation , Epididymis/drug effects , Epididymis/metabolism , Gene Expression/drug effects , Male , Plant Leaves/chemistry , Rats , Rats, Wistar , Reproduction/genetics , Spermatozoa/drug effects , Testis/drug effects , Testis/metabolism
5.
J Ethnopharmacol ; 229: 97-103, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30195059

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Polycystic Ovarian Syndrome (PCOS) is a complex endocrine and reproductive disorder. A main hallmark includes increased androgen production. The root of Paeonia lactiflora Pall. (Bai Shao) is used in Chinese herbal medicine for reproductive disorders, however its effects and mechanisms on ovarian theca cells has not yet been fully elucidated. AIM OF THE STUDY: The aim of this study was to evaluate effect of paeoniflorin extract (PFE), the main constituents of Bai Shao, on androgen production in ovarian theca cells. MATERIALS AND METHODS: Primary murine theca cells were treated with concentrations of PFE (1-100 µg/mL) in the presence of dexamethasone (10 µM) with media-only treated cells used as the control. After 24 h, culture media was collected for biochemistry assays of testosterone and progesterone. Expression of key steroidogenic enzymes, cholesterol side-chain cleavage (CYP11A1) and 17α-hydroxylase (CYP17A1) was characterized using immunofluorescence staining, immunoblotting and qRT-PCR. RESULTS: Dexamethasone significantly enhanced testosterone secretion (P < 0.05 vs. the control cells). PFE reversed over-production of testosterone induced by dexamethasone in a dose-dependent manner. The treatment with PFE also normalized production of progesterone in dexamethasone-treated cells. Expression of CYP11A1 and CYP17A1 in the theca cells were visualised by immunofluorescence staining. All doses of PFE significantly inhibited CYP17A1 expression detected by immunoblotting, but only 100 µg/mL of PFE downregulated CYP11A1 expression and reduced CYP11A1 significantly in dexamethasone-treated theca cells. CONCLUSIONS: PFE may reduce over-secretion of testosterone in theca cells through downregulation of CYP17A1 and CYP11A1. These findings provide scientific evidence to treat ovarian hyperandrogenism with the root of Paeonia lactiflora Pall.


Subject(s)
Glucosides/pharmacology , Monoterpenes/pharmacology , Testosterone/metabolism , Theca Cells/drug effects , Animals , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Dexamethasone , Down-Regulation , Female , Mice , Polycystic Ovary Syndrome , Steroid 17-alpha-Hydroxylase/genetics , Theca Cells/metabolism
6.
Reprod Toxicol ; 82: 25-31, 2018 12.
Article in English | MEDLINE | ID: mdl-30268827

ABSTRACT

Glyphosate has been suggested to be an endocrine disrupting chemical capable of disrupting male reproduction. There are conflicting data, however, with studies reporting effects from exposure to either glyphosate alone or to herbicide formulations, making comparisons difficult. We assessed rat testis histopathology and androgen function following two weeks exposure to either glyphosate at 2.5 and 25 mg/kg bw/day (5x and 50x Acceptable Daily Intake, ADI, respectively), or equivalent high dose of glyphosate in a herbicide formulation; Glyfonova. We observed no significant effects on testes or testosterone synthesis in rats exposed to glyphosate. Limited effects were observed in rats exposed to Glyfonova, with a small upregulation of the steroidogenic genes Cyp11a1 and Cyp17a1. We conclude that glyphosate alone has no effect on adult rat testis at exposure levels up to 25 mg/kg bw/day. Glyfonova induced only minor effects on steroidogenic gene expression, likely caused by additives other than glyphosate.


Subject(s)
Glycine/analogs & derivatives , Herbicides/toxicity , Testis/drug effects , Animals , Cholesterol Side-Chain Cleavage Enzyme/genetics , Glycine/toxicity , Male , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/genetics , Testis/metabolism , Testosterone/metabolism , Glyphosate
7.
Biomed Pharmacother ; 102: 855-864, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29710542

ABSTRACT

This study aimed to determine the protective effects of co-administration of Quercetin (QT) or l-Carnitine (LC) against the oxidative stress induced by Atrazine (ATZ) in the reproductive system of intact male Albino rats. 36 rats were divided equally into 6 groups. Rats in the control negative "CNT" group received 1.5 ml distilled water for 21 days. All rats in the other groups received ATZ (120 mg/kg bw) through gavage. Groups 3 and 4 were co-administered with either low or high dose of QT (10 "ATZLQT" and 50 "ATZHQT" mg/kg bw, respectively). Groups 5 and 6 were co-administered with either low or high dose of LC (200 "ATZLLC" and 400 "ATZHLC" mg/kg bw, respectively). At the end of the experiment, animals were sacrificed and all samples were collected. ATZ significantly increased serum level of malondialdehyde (MDA) and decreased total antioxidant capacity (TAC). Also, ATZ increased significantly the sperm cell abnormalities and reduced both testicular IgA and serum testosterone levels. Testicular DNA laddering % and CYP17A1 mRNA expression were significantly reduced in ATZ group. Interestingly, co-administration with low dose QT or different doses of LC succeeded to counteract the negative toxic effects of ATZ on serum oxidative stress indicators, serum testosterone levels, testicular IgA level and improved testicular CYP17A1 mRNA expression. In conclusion, QT in low dose and LC in both low and high doses exerted a significant protective action against the reproductive toxicity of ATZ, while higher dose of QT failed induce immune-stimulant effect against ATZ in adult male Albino rats.


Subject(s)
Atrazine/toxicity , Carnitine/pharmacology , Quercetin/pharmacology , Reproduction/drug effects , Toxicity Tests , Animals , Antioxidants/metabolism , Body Weight/drug effects , Carnitine/administration & dosage , DNA Fragmentation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Immunoglobulin A/blood , Male , Oxidants/metabolism , Quercetin/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testis/drug effects , Testis/pathology , Testosterone/blood
8.
Anim Sci J ; 88(1): 63-71, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27128796

ABSTRACT

Gossypol, a polyphenolic aldehyde found in cottonseed, has been shown to perturb steroidogenesis in granulosa and luteal cells of rats, pigs and cattle. However, little is known about the direct effect of gossypol on theca cell functions in any species. The present study was conducted to investigate the effect of gossypol on the steroidogenesis and the expression of genes involved in it in cultured bovine theca cells. Theca cells were isolated from healthy preovulatory follicles and were cultured in the presence of luteinizing hormone (LH) for up to 7 days. During the culture period, main steroid products of the theca cells shifted from androstenedione (A4) at day 1 to progesterone (P4) from day 2 onward. At days 1 and 7, theca cells were treated with gossypol (0-25 µg/mL) for 24 h. Gossypol inhibited LH-stimulated theca cell A4 and P4 production in a dose-dependent manner at both occasions. The viability of theca cells was not affected by gossypol at any doses used. Gossypol down-regulated expressions of steroidogenic enzymes CYP11A1, HSD3B1 and CYP17A1, but not that of LHR. These results indicate that gossypol inhibits thecal steroidogenesis through down-regulating gene expressions of steroidogenic enzymes but without affecting cell viability in cattle.


Subject(s)
Androstenedione/biosynthesis , Gossypol/pharmacology , Luteinizing Hormone/pharmacology , Progesterone/biosynthesis , Theca Cells/metabolism , Animals , Cattle , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cottonseed Oil , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Gene Expression/drug effects , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Steroid Isomerases/genetics , Steroid Isomerases/metabolism , Theca Cells/enzymology
9.
Mol Med Rep ; 15(1): 339-344, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27959413

ABSTRACT

Congenital adrenal hyperplasia (CAH) is a rare autosomal recessive disorder caused by mutations in the cytochrome P450 family 17 subfamily A member 1 (CYP17A1) gene located on chromosome 10q24.3, which leads to a deficiency in 17α­hydroxylase/17,20­lyase. The disorder is characterized by low blood levels of estrogens, androgens and cortisol, which leads to a compensatory increase in adrenocorticotropic hormone levels that stimulate the production of mineralocorticoid precursors. This subsequently leads to hypertension, hypokalemia, primary amenorrhea and sexual infantilism. Over 90 distinct genetic lesions have been identified in patients with this disorder. The prevalence of common mutation of CYP17A1 gene differs among ethnic groups. Treatment of this disorder involves replacement of glucocorticoids and sex steroids. Estrogen alone is prescribed for patients who are biologically male with 17α­hydroxylase deficiencies that identify as female. However, genetically female patients may receive estrogen and progesterone supplementation. In the present study, a 17­year­old female with 17α­hydroxylase/17,20­lyase deficiency that presented with primary amenorrhea and sexual infantilism and no hypertension, was examined. The karyotype of the patient was 46, XX, and genetic analysis revealed the presence of a compound heterozygous mutation in exons 6 and 8, leading to the complete absence of 17α­hydroxylase/17,20­lyase activity. The patient was treated with prednisolone and ethinyl estradiol. In addition, a summary of the recent literature regarding CAH is presented.


Subject(s)
Adrenal Hyperplasia, Congenital/complications , Adrenal Hyperplasia, Congenital/genetics , Steroid 17-alpha-Hydroxylase/genetics , Adolescent , Adrenal Hyperplasia, Congenital/drug therapy , Amenorrhea/complications , Amenorrhea/drug therapy , Amenorrhea/genetics , Estrogens/therapeutic use , Ethinyl Estradiol/therapeutic use , Exons , Female , Glucocorticoids/therapeutic use , Humans , Karyotype , Mutation , Prednisolone/therapeutic use , Sexual Infantilism/complications , Sexual Infantilism/drug therapy , Sexual Infantilism/genetics
10.
Reprod Toxicol ; 66: 107-114, 2016 12.
Article in English | MEDLINE | ID: mdl-27773742

ABSTRACT

Isoliquiritigenin is a botanical estrogen used as a dietary supplement. Previous studies show that other botanical estrogens affect ovarian estradiol synthesis, but isoliquiritigenin's effects on the ovary are unknown. Thus, this study tested the hypothesis that isoliquiritigenin inhibits ovarian antral follicle growth and steroidogenesis. Antral follicles from CD-1 mice were cultured with vehicle control (dimethyl sulfoxide; DMSO) or isoliquiritigenin (0.6µM, 6 µM, 36 µM, and 100 µM) for 48-96h. During culture, follicle diameters were measured daily to assess follicle growth. After culture, media were collected for hormone assays and follicles were collected for gene expression analysis of steroidogenic enzymes. Isoliquiritigenin inhibited antral follicle growth and altered estradiol, testosterone, and progesterone levels. Additionally, isoliquiritigenin altered the mRNA levels of cytochrome P450 steroid 17-α-hydroxylase 1, aromatase, 17ß-hydroxysteroid dehydrogenase 1, and steroidogenic acute regulatory protein. These data indicate that exposure to isoliquiritigenin inhibits growth and disrupts steroid production in antral follicles.


Subject(s)
Chalcones/toxicity , Ovarian Follicle/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Aromatase/genetics , Estradiol/metabolism , Female , Mice , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Phosphoproteins/genetics , Progesterone/metabolism , RNA, Messenger/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Testosterone/metabolism
11.
Pharm Biol ; 54(6): 975-83, 2016.
Article in English | MEDLINE | ID: mdl-26459667

ABSTRACT

CONTEXT: Quercetin, a flavonoid, has been tried in traditional medicine for treating many disorders and reported to have inhibitory action on PI3 kinase. OBJECTIVE: This study investigates the effect of quercetin on testosterone propionate induced polycystic ovary syndrome (PCOS) model, which shows both metabolic and endocrine features of PCOS. MATERIALS AND METHODS: Female pre-pubertal Sprague-Dawley rats were randomly divided into four groups: normal control, PCOS control, quercetin, and metformin treated. PCOS was induced by testosterone propionate (10 mg/kg, s.c.) and treatments were carried out orally at the dose of 150 mg/kg from the 6th week. At the 6th and 10th week, blood was collected to investigate metabolic indices, and reproductive biochemical parameters including morphology of ovary, uterus, and estrous cyclicity were assessed. The ovaries were processed to determine CYP17A1 gene expression. RESULTS: The treatment with quercetin did not modify body weight gain but uterine (296.7 ± 5.11 versus 263.0 ± 8.60 mg) and ovary weights (49.5 ± 1.93 versus 37.8 ± 3.43 mg) were found to be decreased significantly (p <0.05) as compared with the PCOS control group. The PCOS control group showed hyperinsulinemia, hyperandrogenemia, and dyslipidemia. Treatment with quercetin showed statistically significant (p <0.01) improvement in insulin (12.46 ± 0.3 versus 10.0 ± 0.28 µU/ml), testosterone (0.65 ± 0.02 versus 0.29 ± 0.02 µU/ml), luteinising hormone (20.6 ± 0.28 versus 15.1 ± 0.36 U/ml), and lipid profile. Histological examination of ovary and uterus confirmed the disease occurrence and remission state in the diseased and treated groups, respectively. Quercetin also demonstrated PI3 kinase inhibition in a docking study and decreased CYP17A1 gene expression. DISCUSSION AND CONCLUSION: Thus, we can conclude that quercetin may have beneficial effect in PCOS by virtue of inhibition of PI3K which attributes to a decrease in the expression of CYP17A1 gene, having a key role in steroidogenesis.


Subject(s)
Ovary/drug effects , Phosphoinositide-3 Kinase Inhibitors , Polycystic Ovary Syndrome/drug therapy , Quercetin/therapeutic use , Steroid 17-alpha-Hydroxylase/genetics , Animals , Body Weight/drug effects , Disease Models, Animal , Estrous Cycle/drug effects , Female , Gene Expression/drug effects , Molecular Docking Simulation , Organ Size/drug effects , Ovary/metabolism , Ovary/pathology , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/genetics , Protein Binding , Quercetin/administration & dosage , Rats, Sprague-Dawley , Uterus/drug effects , Uterus/metabolism , Uterus/pathology
12.
Reprod Toxicol ; 57: 130-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26055946

ABSTRACT

Seafood products, including fish and fish oils, are major sources of persistent organic pollutants (POPs) which may cause endocrine disruption related to reproductive dysfunction in males. Primary porcine neonatal Leydig cells were exposed to three extracts of POPs obtained from different stages in production of cod liver oil dietary supplement, in the absence and presence of luteinizing hormone (LH). No reduced viability was observed and all POP extracts showed increased testosterone and estradiol levels in unstimulated cells and decreased testosterone and estradiol secretion in LH-stimulated cells. A decrease in central steriodogenic genes including STAR, CYP11A1, HSD3B and CYP17A1 was obtained in both culture conditions with all POP extracts. We implicate both small differences in composition and concentration of compounds as well as "old" POPs to be important for the observed steroidogenic effects.


Subject(s)
Cod Liver Oil/chemistry , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Hydrocarbons, Chlorinated/toxicity , Leydig Cells/drug effects , Animals , Animals, Newborn , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Estradiol/metabolism , Gene Expression/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Male , Multienzyme Complexes/genetics , Phosphoproteins/genetics , Progesterone Reductase/genetics , Steroid 17-alpha-Hydroxylase/genetics , Steroid Isomerases/genetics , Swine , Testosterone/metabolism
13.
Reprod Toxicol ; 57: 100-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26049154

ABSTRACT

Effects of triphenyl phosphate (TPP) and tris-(2-chloroethyl) phosphate (TCEP) exposure on induction of oxidative stress and endocrine disruption were investigated in TM3 cells. After 24h exposure, cell growth declined and morphology changed in TPP and TCEP treated groups with high dosages. Significant increases in superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX) and glutathione S-transferase (GST) activities and their respective gene expressions in a dose-dependent and/or time-dependent manner in TPP or TCEP groups. Moreover, the expression of main genes related to testosterone (T) synthesis including cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), cytochrome P450 17α-hydroxysteroid dehydrogenase (P450-17α), 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 17ß-hydroxysteroid dehydrogenase (17ß-HSD) were dramatically reduced by TPP and TCEP treatments, especially with the high dosage for 24h. TPP and TCEP treatments for 24h caused significant decreases in T levels in the medium. Furthermore, co-treatments of hCG with TPP or TCEP could inhibit hCG-induced changes in the expression of P450scc, P450-17α and 17ß-HSD and T levels. Taken together, TPP and TCEP could induce oxidative stress and endocrine disruption in TM3 cells.


Subject(s)
Leydig Cells/drug effects , Organophosphates/toxicity , Phosphines/toxicity , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Catalase/genetics , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Gene Expression/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Leydig Cells/metabolism , Male , Mice , Oxidative Stress/drug effects , Phosphoproteins/genetics , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, LDL/genetics , Scavenger Receptors, Class B/genetics , Steroid 17-alpha-Hydroxylase/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Testosterone/metabolism
14.
Clinics (Sao Paulo) ; 70(2): 144-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25789524

ABSTRACT

OBJECTIVE: To analyze steroidogenesis-related gene expression in the rat ovary exposed to melatonin supplementation. METHODS: Thirty-two virgin adult female rats were randomized to two groups as follows: the control group GI received vehicle and the experimental group GII received melatonin supplementation (10 µg/night per animal) for 60 consecutive days. After the treatment, animals were anesthetized and the collected ovaries were immediately placed in liquid nitrogen for complementary deoxyribonucleic acid microarray analyses. A GeneChip(®) Kit Rat Genome 230 2.0 Affymetrix Array was used for gene analysis and the experiment was repeated three times for each group. The results were normalized with the GeneChip(®) Operating Software program and confirmed through analysis with the secondary deoxyribonucleic acid-Chip Analyzer (dChip) software. The data were confirmed by real-time reverse transcription polymerase chain reaction analysis. Genes related to ovarian function were further confirmed by immunohistochemistry. RESULTS: We found the upregulation of the type 9 adenylate cyclase and inhibin beta B genes and the downregulation of the cyclic adenosine monophosphate response element modulator and cytochrome P450 family 17a1 genes in the ovarian tissue of GII compared to those of the control group. CONCLUSION: Our data suggest that melatonin supplementation decreases gene expression of cyclic adenosine monophosphate, which changes ovarian steroidogenesis.


Subject(s)
Adenylyl Cyclases/genetics , Gene Expression/drug effects , Inhibin-beta Subunits/genetics , Melatonin/pharmacology , Ovary/drug effects , Adenylyl Cyclases/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/metabolism , Dietary Supplements , Female , Inhibin-beta Subunits/metabolism , Melatonin/metabolism , Models, Animal , Ovary/metabolism , RNA, Complementary/isolation & purification , Random Allocation , Rats, Wistar , Real-Time Polymerase Chain Reaction/methods , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Tissue Array Analysis/methods , Up-Regulation
15.
J Physiol Sci ; 65(3): 253-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25715777

ABSTRACT

We investigated age-induced changes in mRNA expression profiles of sex-steroidogenic enzymes and sex-steroid receptors in 3-, 12-, and 24-month-old male rat brain subregions [cerebral cortex (CC), hypothalamus (Hy) and cerebellum (CL)]. In many cases, the expression levels of mRNA decreased with age for androgen synthesis enzyme systems, including Cyp17a1, Hsd17b and Srd5a in the CC and CL, but not in the Hy. Estradiol synthase Cyp19a1 did not show age-induced decline in the Hy, and nearly no expression of Cyp19a1 was observed in the CC and CL over 3-24 m. Androgen receptor Ar increased in the Hy but decreased in the CC with age. Estrogen receptor Esr1 increased in the CC and Hy, and did not change in the CL with age. Esr2 did not change in the CC and Hy, but decreased in the CL with age. As a comparison, age-induced changes of brain-derived neurotrophic factor mRNA were also investigated.


Subject(s)
Aging/metabolism , Brain/metabolism , Gonadal Steroid Hormones/biosynthesis , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Aging/genetics , Animals , Aromatase/genetics , Aromatase/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Hypothalamus/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
16.
Gen Comp Endocrinol ; 221: 134-43, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25701739

ABSTRACT

Estradiol-17ß (E2) and maturation-inducing hormone (MIH) are two steroid hormones produced in the teleost ovary that are required for vitellogenic growth and final oocyte maturation and ovulation. During this transition, the main steroid hormone produced in the ovary shifts from estrogens to progestogens. In the commercially important Japanese eel (Anguilla japonica), the MIH 17α,20ß-dihydroxy-4-pregnen-3-one (DHP) is generated from its precursor by P450c17, which has both 17α-hydroxylase and C17-20 lyase activities. In order to elucidate the regulatory mechanism underlying the steroidogenic shift from E2 to DHP and the mechanistic basis for the failure of this shift in artificially matured eels, the cDNA for cyp17a2-which encodes P450c17-II-was isolated from the ovary of wild, mature Japanese eel and characterized, and the expression patterns of cyp17a1 and cyp17a2 during induced ovarian development were investigated in cultured eel ovaries. Five cDNAs (types I-V) encoding P450c17-II were identified that had minor sequence variations. HEK293T cells transfected with all but type II P450c17-II converted exogenous progesterone to 17α-hydroxyprogesterone (17α-P), providing evidence for 17α-hydroxylase activity; however, a failure to convert 17α-P to androstenedione indicated that C17-20 lyase activity was absent. Cyp17a2 mRNA was expressed mainly in the head kidney, ovary, and testis, and quantitative PCR analysis demonstrated that expression in the ovary increased during induced vitellogenesis and oocyte maturation/ovulation. In contrast, P450c17-I showed both 17α-hydroxylase and C17-20 lyase activities, and cyp17a1 expression increased until the mid-vitellogenic stage and remained high thereafter. Considering the high level of cyp17a2 transcript in the eel ovary at the migratory nucleus stage together with our previous report demonstrating that eel ovaries have strong 17α-P-to-DHP conversion activity, the failure of artificially maturing eels to produce the maturation-inducing DHP may be explained by a deficiency in 17α-P production due to the persistence of cyp17a1 expression after the completion of vitellogenesis.


Subject(s)
Anguilla/metabolism , DNA, Complementary/genetics , Gene Expression Regulation, Developmental/physiology , Ovary/embryology , Steroid 17-alpha-Hydroxylase/genetics , Vitellogenesis/physiology , Amino Acid Sequence , Anguilla/genetics , Anguilla/growth & development , Animals , Base Sequence , Estradiol/metabolism , Female , HEK293 Cells , Humans , Male , Molecular Sequence Data , Progesterone/metabolism , Progestins/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sex Factors , Steroid 17-alpha-Hydroxylase/metabolism , Steroids/metabolism , Testis/metabolism
17.
J Physiol Pharmacol ; 65(1): 127-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24622837

ABSTRACT

Phytoestrogens (PEs), including genistein and daidzein, are plant-derived substances that mimic or antagonize estrogen action in animals. The majority of studies investigated the effects of PEs on reproduction in humans and laboratory animals. The mechanisms of phytoestrogen action on reproductive processes in domesticated animals, including pigs, are garnering increasing attention. However, very few in vivo and in vitro studies investigating the effects of PEs on adrenal glands have been carried out on models other than humans and rats. The aim of the present study was to determine whether the effects of genistein and daidzein on adrenal in vitro steroidogenesis are accompanied by changes in expression of genes encoding key steroidogenic enzymes in porcine adrenocortical cells. The following genes were analyzed: cholesterol side-chain cleavage enzyme (P450scc, CYP11A1 gene), 3ß-hydroxysteroid dehydrogenase (3ß-HSD, HSD3B1 gene), 17α-hydroxylase/C17-20 lyase (P450c17, CYP17A1 gene) and 21-hydroxylase (P450c21, CYP21A2 gene). Porcine adrenocortical cells collected from both luteal- and follicular-phase gilts were exposed for eight hours to genistein (10 µM), or daidzein (10 µM), in the absence or presence of ACTH (5 nM). Genistein and daidzein inhibited basal and ACTH-stimulated secretion of cortisol and corticosterone and stimulated secretion of androstenedione. PEs did not affect the expression of CYP11A1, HSD3B1, CYP17A1 and CYP21A2 in the adrenocortical cells of luteal- and follicular-phase gilts. It can be concluded that the influence of PEs on steroid secretion in porcine adrenal glands is not mediated by changes in the expression of genes encoding major steroidogenic enzymes. More studies are needed to elucidate the intracellular mechanisms leading to the PE-induced changes in adrenal steroidogenesis in pigs.


Subject(s)
Adrenal Cortex/metabolism , Androstenedione/metabolism , Corticosterone/metabolism , Genistein/pharmacology , Hydrocortisone/metabolism , Isoflavones/pharmacology , 3-Hydroxysteroid Dehydrogenases/genetics , Adrenal Cortex/cytology , Animals , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Female , Follicular Phase/metabolism , Gene Expression/drug effects , Luteal Phase/metabolism , Phytoestrogens/pharmacology , Steroid 17-alpha-Hydroxylase/genetics , Steroid 21-Hydroxylase/genetics , Swine
18.
PLoS One ; 8(11): e79686, 2013.
Article in English | MEDLINE | ID: mdl-24223995

ABSTRACT

A new role for fat supplements, in particular conjugated linoleic acid (CLA), has been delineated in steroidogenesis, although the underlying molecular mechanisms have not yet been elucidated. The aims of the present study were to identify the pathway stimulated by CLA supplementation using a cell culture model and to determine whether this same pathway is also stimulated in vivo by CLA supplementation associated with exercise. In vitro, Leydig tumour rat cells (R2C) supplemented with different concentrations of CLA exhibited increasing testosterone biosynthesis accompanied by increasing levels of CYP17A1 mRNA and protein. In vivo, trained mice showed an increase in free plasma testosterone and an up-regulation of CYP17A1 mRNA and protein. The effect of training on CYP17A1 expression and testosterone biosynthesis was significantly higher in the trained mice supplemented with CLA compared to the placebo. The results of the present study demonstrated that CLA stimulates testosterone biosynthesis via CYP17A1, and endurance training led to the synthesis of testosterone in vivo by inducing the overexpression of CYP17A1 mRNA and protein in the Leydig cells of the testis. This effect was enhanced by CLA supplementation. Therefore, CLA-associated physical activity may be used for its steroidogenic property in different fields, such as alimentary industry, human reproductive medicine, sport science, and anti-muscle wasting.


Subject(s)
Dietary Supplements , Linoleic Acids, Conjugated/pharmacology , Physical Conditioning, Animal , Physical Endurance , Steroid 17-alpha-Hydroxylase/metabolism , Testosterone/biosynthesis , Up-Regulation/drug effects , Animals , Cell Line, Tumor , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Steroid 17-alpha-Hydroxylase/genetics
19.
PLoS One ; 8(11): e79382, 2013.
Article in English | MEDLINE | ID: mdl-24260211

ABSTRACT

Hyperandrogenism is a core factor in the series of reproductive and endocrine metabolic disorders involved in polycystic ovary syndrome (PCOS). Abnormalities in enzymatic activity and the expression of ovarian granular cell layer P450arom and theca cell P450c17α can lead to an atypical environment of local ovarian hormones, including excessive androgen levels. Rat models prepared with letrozole exhibit similar endocrine and histological changes to those that occur in human PCOS. We used such a model to study the role of electro-acupuncture (EA) in regulating ovarian P450arom and P450c17α enzymatic activity and mRNA expression in PCOS rats. Female Sprague Dawley (SD) rats aged 42 days were randomly divided into 3 groups (control, PCOS, and PCOS EA) consisting of 10 rats each. The PCOS and PCOS EA groups were administered a gavage of 1.0 mg/kg(-1) of letrozole solution once daily for 21 consecutive days. Beginning in the ninth week, the PCOS EA group was administered low-frequency EA treatment daily for 14 consecutive days. After the treatment, we obtained the following results. The estrous cycles were restored in 8 of the 10 rats in the PCOS EA group, and their ovarian morphologies and ultrastructures normalized. The peripheral blood measurements (with ELISA) showed significantly decreased androgens (i.e., androstenedione and testosterone) with significantly increased estrogens (i.e., estrone, estradiol) and increased P450arom with decreased P450C17α. Immunohistochemistry and Western blotting methods showed enhanced expression of ovarian granular cell layer P450arom as well as decreased expression of theca cell layer P450C17α. Fluorescence quantitative PCR methods showed enhanced expression of ovarian granular cell layer P450arom mRNA as well as decreased expression of theca cell layer P450C17α mRNA. These results may help explain the effects of electro-acupuncture in changing the local ovarian hyperandrogenic environment and improving reproductive and endocrine metabolic disorders in PCOS.


Subject(s)
Acupuncture Therapy/methods , Aromatase/metabolism , Nitriles/pharmacology , Ovary/drug effects , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Triazoles/pharmacology , Animals , Aromatase/genetics , Female , Immunohistochemistry , Letrozole , Microscopy, Electron, Transmission , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/genetics
20.
Biol Res ; 46(2): 139-46, 2013.
Article in English | MEDLINE | ID: mdl-23959011

ABSTRACT

Studies of developmental effects of mixtures of endocrine disrupters on the male reproductive system are of great concern. In this study, the reproductive effects of the co-administration of di-2-(ethylhexyl) phthalate (DEHP) and genistein (GEN) during pregnancy and lactation were studied in male rat offspring. Pregnant Sprague-Dawley rats were gavaged from gestation day 3 to postnatal day 21 with vehicle control, DEHP 250 mg/kg body weight (bwyday, GEN 50 mg/kg bwday, GEN 400 mg/kg bwday, and two combinations of the two compounds (DEHP 250 mg/kg bwday + GEN 50 mg/kg bwday, DEHP 250 mg/kg bwday + GEN 400 mg/kg bwday). The outcomes studied were general morphometry (weight, AGD), testicular histology, testosterone levels, and expression at the mRNA level of genes involved in steroidogenesis. Organ coefficient, AGD / body weight1/3 י, serum testosterone concentration and genes involved in steroidogenic pathway expression when exposed to DEHP (250mg/kg bwday), GEN(50mg/kg bwday) or GEN(400mg/kg bwday) alone were not significantly different from the control group. When exposed to (DEHP 250mg/kg bwday +GEN 50mg/kg bwday) together during pregnancy and lactation, serum testosterone concentration, epididymis coefficient and Cypal17a1,Scarb1 m RNA expression significantly decreased compared to the control and GEN(50mg/kg bwday). When exposed to (DEHP 250mg/kg bwday +GEN 400mg/kg bwday) together during pregnancy and lactation, AGD / body weight1/3 י, serum testosterone concentration, testis and epididymis coefficient and Star, Cypal17a1 mRNA expression appeared significantly decreased compared to the control and DEHP/GEN single exposure, together with developmental impairment of seminiferous tubules and seminiferous epithelium. Overall, co-administration of DEHP and GEN during gestation and lactation seem to acts in a cumulative manner to induce the most significant alterations in the neonate, especially with GEN at high dose, although the effect of the DEHP-GEN mixture on adult offspring should be observed further.


Subject(s)
Diethylhexyl Phthalate/toxicity , Endocrine Disruptors/toxicity , Genistein/toxicity , Genitalia, Male/drug effects , Lactation/drug effects , Phytoestrogens/toxicity , Plasticizers/toxicity , Animals , Cytochrome P-450 CYP11B2/genetics , Female , Male , Maternal Exposure/adverse effects , Phosphoproteins/genetics , Pregnancy , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B/genetics , Steroid 17-alpha-Hydroxylase/genetics , Testis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL