Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Endocr J ; 68(10): 1165-1177, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-33980773

ABSTRACT

The toxicity of certain novel perfluoroalkyl substances (PFCs) has attracted increasing attention. However, the toxic effects of sodium p-perfluorous nonenoxybenzene sulfonate (OBS) on the endocrine system have not been elucidated. In this study, OBS was added to the drinking water during the pregnancy and lactation of the healthy female mice at dietary levels of 0.0 mg/L (CON), 0.5 mg/L (OBS-L), and 5.0 mg/L (OBS-H). OBS exposure during the pregnancy and lactation resulted in the presence of OBS residues in the placenta and fetus. We also analyzed physiological and biochemical parameters and gene expression levels in mice of the F0 and F1 generations after maternal OBS exposure. The total serum cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels were significantly increased in female mice of the F0 generation. The androgen levels in the serum and the ovarian mRNA levels of androgen receptor (AR) also tended to increase after maternal OBS exposure in the F0 generation mice. Moreover, maternal OBS exposure altered the mRNA expression of endocrine-related genes in male mice of F1 generation. Notably, the serum TC and LDL-C levels were significantly increased in 8-weeks-old male mice of the F1 generation, and the serum high-density lipoprotein cholesterol (HDL-C) levels were decreased in 24-week-old male mice of the F1 generation. These results indicated that maternal OBS exposure can interfere with endocrine homeostasis in the F0 and F1 generations. Therefore, exposure to OBS during pregnancy and lactation has the potential toxic effects on the dams and male offspring, which cannot be overlooked.


Subject(s)
Endocrine Disruptors/toxicity , Estrogen Receptor alpha/drug effects , Maternal Exposure , Ovary/drug effects , Receptors, Androgen/drug effects , Testis/drug effects , Uterus/drug effects , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/blood , Animals , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Estrogen Receptor alpha/genetics , Estrogens/blood , Female , Fetus/chemistry , Lactation , Male , Mice , Organ Size , Ovary/pathology , Placenta/chemistry , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Testis/chemistry , Testis/pathology , Uterus/chemistry , Uterus/pathology
2.
J Nat Prod ; 83(10): 3080-3092, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33021790

ABSTRACT

There are no targeted therapies available for triple-negative breast cancers (TNBCs) in part because they represent a heterogeneous group of tumors with diverse oncogenic drivers. Our goal is to identify targeted therapies for subtypes of these cancers using a mechanism-blind screen of natural product extract libraries. An extract from Desmanthodium guatemalense was 4-fold more potent for cytotoxicity against MDA-MB-231 cells, which represent the mesenchymal stem-like (MSL) subtype, as compared to cells of other TNBC subtypes. Bioassay-guided fractionation led to the isolation of six polyacetylenes, and subsequent investigations of plant sources known to produce polyacetylenes yielded six additional structurally related compounds. A subset of these compounds retained selective cytotoxic effects in MSL subtype cells. Studies suggest that these selective effects do not appear to be due to PPARγ agonist activities that have previously been reported for polyacetylenes. A CRISPR-Cas9-mediated gene knockout screen was employed to identify the mechanism of selective cytotoxic activity of the most potent and selective compound, dehydrofalcarinol (1a). This genomic screen identified HSD17B11, the gene encoding the enzyme 17ß-hydroxysteroid dehydrogenase type 11, as a mediator of the selective cytotoxic effects of 1a in MDA-MB-231 cells that express high levels of this protein. The Project Achilles cancer dependency database further identified a subset of Ewing sarcoma cell lines as highly dependent on HSD17B11 expression, and it was found these were also highly sensitive to 1a. This report demonstrates the value of CRISPR-Cas9 genome-wide screens to identify the mechanisms underlying the selective activities of natural products.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , CRISPR-Cas Systems , Gene Knockout Techniques/methods , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/drug therapy , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , Aldehyde Oxidoreductases/drug effects , Aldehyde Oxidoreductases/genetics , Cell Line, Tumor , Female , Humans , Molecular Structure , PPAR gamma/agonists , RNA, Small Interfering/pharmacology
3.
Technol Health Care ; 27(S1): 357-365, 2019.
Article in English | MEDLINE | ID: mdl-31045553

ABSTRACT

Dexamethasone (DEX) is associated with many inflammation and metabolic diseases. We analyzed the effects of DEX on the expression of estrogen metabolism enzyme 17ß-HSD1 at the optic nerve. Rats were treated with different concentrations of intraperitoneal DEX. Western Blot analysis showed that 17ß-HSD protein was expressed in the optic nerve tissue. The enzyme was detected by immunohistochemistry on the terminal foot of Muller cells from the ganglion cell layer of rat retina. ELISA analysis showed that the 17ß-HSD1 protein expression of DEX-treated group is 2.4 fold comparing to the control group. The results indicated that DMXS sodium phosphate might modulate the expression of 17ß-HSD1 protein in optic tissue. This study sheds light on understanding of the relationship among DEX, 17ß-HSD presence and distribution of visual neural systems. At the same time, DEX treatment affects the athletic ability and memory of the animals. Compared with the control group, the experimental group showed slow response to stimulation, inertia, depression, cowardice and lack of appetite. The results of ethology experiments showed that all the parameters decreased by 15-30%.


Subject(s)
17-Hydroxysteroid Dehydrogenases/drug effects , Dexamethasone/pharmacology , Optic Nerve/drug effects , Animals , Blotting, Western , Dexamethasone/administration & dosage , Enzyme-Linked Immunosorbent Assay , Rats , Rats, Sprague-Dawley
4.
J Anim Sci ; 97(2): 851-864, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30508170

ABSTRACT

The aim of this study was to investigate the effect of orexin B (OXB) on progesterone (P4) and androstenedione (A4) secretion by porcine endometrial and myometrial tissue explants and on the expression of key steroidogenic proteins and enzymes involved in steroid production. The hormones secretion and the expression of steroidogenic acute regulatory protein (StAR), P450 side-chain cleavage enzyme (CYP11A1), and 3ß-hydroxysteroid dehydrogenase (HSD3B1) were analyzed on days 10 to 11, 12 to 13, 15 to 16, and 27 to 28 of pregnancy and during the luteal phase of the estrous cycle (days 10 to 11). Endometrial and myometrial explants were cultured in vitro in the presence of OXB (1, 10, or 100 nM) and OXB (1, 10, or 100 nM) with 1 µM of JNJ (OX2R antagonist). Gene expression was examined by real-time PCR, and steroid secretion was determined by radioimmunoassay. Orexin B modulated StAR, CYP11A1, HSD3B1 mRNA content depending on the type of uterine tissue, the applied OXB dose, and the stage of pregnancy or the estrous cycle (P < 0.05). Orexin B increased P4 secretion in all stages of early gestation (P < 0.05). Orexin B enhanced the release of A4 on days 12 to 13, 15 to 16, and 27 to 28 of gestation, whereas on days 10 to 11 of early pregnancy, A4 secretion decreased in the endometrium and increased in the myometrium (P < 0.05). These results indicate that OXB affects the expression of key steroidogenic regulators and the secretion of steroid hormones in the porcine uterus during early pregnancy.


Subject(s)
17-Hydroxysteroid Dehydrogenases/drug effects , Gene Expression Regulation/drug effects , Orexins/pharmacology , Phosphoproteins/drug effects , Reproduction , Swine/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Androstenedione/metabolism , Animals , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/genetics , Endometrium/metabolism , Estrous Cycle/drug effects , Female , Myometrium/metabolism , Phosphoproteins/genetics , Pregnancy , Progesterone/metabolism , RNA, Messenger/metabolism , Swine/genetics , Uterus/metabolism
5.
Chemosphere ; 216: 94-102, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30359922

ABSTRACT

Natural and synthetic progestins in receiving streams can disrupt the normal endocrine systems of fish. Norethindrone (NET) is a widely used synthetic progestin that often appears in wastewater effluents. For this research, adult female western mosquitofish (Gambusia affinis) were exposed to NET at three concentrations. The effects of NET on the following biological factors were evaluated: the histology of the ovaries and livers, the anal fin morphology, and transcription of genes related to steroidogenesis signaling pathways in the livers. After 42 d exposure to NET at 33.0 ng L-1 and 347.5 ng L-1, rapid masculinization, an increase in the number of atretic and postovulatory follicles in the ovary, enhanced vascularization, degenerated hepatocytes and irregular nuclei in the livers were observed. Exposure to NET did not affect the expression of the androgenic and estrogenic receptor genes and Cyp19a except for a significant up-regulation of Erα. However, the expression of Vtg A, Vtg B, and Vtg C were markedly inhibited in the females exposed to three concentrations of NET. Compared to the control female, exposure to NET at 33.0 ng L-1 and 347.5 ng L-1 caused a 4.4- and 5.8-fold increase in the expression of Hsd17ß3 in the livers, respectively. The results demonstrate that NET can cause rapid masculinization of female G. affinis, hepatopathological alterations and inhibited expressions of Vtg A, Vtg B, and Vtg C. The results imply that G. affinis populations might be threatened in NET-contaminated environment.


Subject(s)
Cyprinodontiformes/anatomy & histology , Liver/drug effects , Norethindrone/pharmacology , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Androgens/metabolism , Animals , Contraceptives, Oral, Synthetic/pharmacology , Cyprinodontiformes/metabolism , Endocrine System/metabolism , Estrogen Receptor alpha/metabolism , Female , Liver/metabolism , Ovary/drug effects , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/pharmacology
6.
Gynecol Endocrinol ; 33(1): 48-52, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27449565

ABSTRACT

Polycystic ovary syndrome (PCOS) is a type of endocrine metabolic disorder with many different consequences to health, most commonly infertility, obesity and insulin resistance. Trivalent chromium (Cr3+) was previously found to improve the metabolic profiles of patients with PCOS. The aim of this study was to explore the effect of Cr on regulating steroidogenic enzymes in adipose tissue. Female BALB/c mice were divided into three groups (n = 6 per group): the control group, PCOS + placebo milk group and PCOS + Cr-containing milk group. The dietary intake of Cr significantly decreased fasting blood sugar (FBS) and homeostasis model assessment of insulin resistance levels in the murine model of PCOS. Importantly, we found significant correlations among the levels of Cr, insulin and dehydroepiandrosterone (DHEA). In adipose tissue, decreases in the enzyme expressions of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 17ß-hydroxysteroid dehydrogenase, but not of aromatase, were observed. By understanding the role of steroidogenic enzymes in PCOS in normal and pathological states, trace elements may be used as a form of adjunctive therapy in the management of patients with PCOS.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Adipose Tissue/metabolism , Aromatase/metabolism , Chromium/pharmacology , Dehydroepiandrosterone/metabolism , Polycystic Ovary Syndrome/drug therapy , Trace Elements/pharmacology , 17-Hydroxysteroid Dehydrogenases/drug effects , Adipose Tissue/drug effects , Animals , Aromatase/drug effects , Chromium/administration & dosage , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Trace Elements/administration & dosage
7.
Asian J Androl ; 18(3): 456-61, 2016.
Article in English | MEDLINE | ID: mdl-26289400

ABSTRACT

This study was to investigate the effect of annexin A5 on testosterone secretion from primary rat Leydig cells and the underlying mechanisms. Isolated rat Leydig cells were treated with annexin A5. Testosterone production was detected by chemiluminescence assay. The protein and mRNA of Steroidogenic acute regulatory (StAR), P450scc, 3ß-hydroxysteroid dehydrogenase (3ß-HSD), 17ß-hydroxysteroid dehydrogenase (17ß-HSD), and 17α-hydroxylase were examined by Western blotting and semi-quantitative RT-PCR, respectively. Annexin A5 significantly stimulated testosterone secretion from rat Leydig cells in dose- and time-dependent manners and increased mRNA and protein expression of StAR, P450scc, 3ß-HSD, and 17ß-HSD but not 17α-hydroxylase. Annexin A5 knockdown by siRNA significantly decreased the level of testosterone and protein expression of P450scc, 3ß-HSD, and 17ß-HSD. The significant activation of ERK1/2 signaling was observed at 5, 10, and 30 min after annexin A5 treatment. After the pretreatment of Leydig cells with ERK inhibitor PD98059 (50 µmol l-1 ) for 20 min, the effects of annexin A5 on promoting testosterone secretion and increasing the expression of P450scc, 3ß-HSD, and 17ß-HSD were completely abrogated (P < 0.05). Thus, ERK1/2 signaling is involved in the roles of annexin A5 in mediating testosterone production and the expression of P450scc, 3ß-HSD, and 17ß-HSD in Leydig cells.


Subject(s)
Annexin A5/pharmacology , Enzyme Inhibitors/pharmacology , Leydig Cells/drug effects , MAP Kinase Signaling System/drug effects , RNA, Messenger/drug effects , Testosterone/biosynthesis , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Blotting, Western , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Leydig Cells/metabolism , Male , Phosphoproteins/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
8.
Andrologia ; 47(5): 560-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24909355

ABSTRACT

The objective of the present study was to investigate the effects of testosterone in recuperation of lead-induced suppressed reproduction in adult male rats. Lead acetate was administered orally to adult male rats (95 ± 5 days) at dosage level of 0.05 and 0.15% for 55 days through drinking water and injected intraperitoneally with either testoviron depot at a dose of 4.16 mg kg(-1) body weight or vehicle alone on days 1, 7 and 14 respectively. At the end of treatment, control and treated males were cohabited with untreated normal-cycling females. After cohabitation for 5 days, all the male rats were killed and weights of reproductive organs were determined. Significant increase in the indices of testis, epididymis, seminal vesicles, vas deferens and prostate glands was observed in testosterone (T)-treated rats when compared to those of lead-exposed rats. Testosterone treatment significantly increased epididymal sperm count, motile spermatozoa, viable spermatozoa and HOS tail-coiled spermatozoa and also the activity levels of testicular 3ß- and 17ß-hydroxysteroid dehydrogenases when compared to those of lead-exposed males. From the results, it can be hypothesised that supplementation of testosterone mitigated lead-induced suppressed reproduction in male rats.


Subject(s)
Androgens/pharmacology , Infertility, Male/chemically induced , Organometallic Compounds/toxicity , Spermatozoa/drug effects , Testis/drug effects , Testosterone/pharmacology , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Epididymis/drug effects , Epididymis/pathology , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Luteinizing Hormone/drug effects , Male , Organ Size , Prostate/drug effects , Prostate/pathology , Rats , Rats, Wistar , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Sperm Motility/drug effects , Testis/metabolism , Testis/pathology , Vas Deferens/drug effects , Vas Deferens/pathology
9.
Andrologia ; 44(2): 92-101, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21933222

ABSTRACT

This study investigated the effects of gestational and lactational exposure to lead and cadmium on testicular steroidogenesis, antioxidant system and male accessory gland functions in F1 generation rats to understand the biochemical mechanisms involved in endocrine disruptions. Pregnant rats were subcutaneously administered with 0.05 mg kg(-1) body wt\ day(-1) of sodium acetate (control), lead acetate, cadmium acetate and (lead acetate + cadmium acetate) throughout the gestational-lactational period, and all animals from each of the experimental groups were sacrificed by decapitation on post-natal day 56 for performing various biochemical assays. We observed significant reduction in the activities of testicular key steroidogenic enzymes and serum testosterone concentration along with significant depletion in cholesterol, ascorbic acid and reduced glutathione contents in all the metal-treated groups. Reductions in the activities of catalase and superoxide dismutase with concomitant increase in the levels of thiobarbituric acid reactive substance were observed in experimental groups. Both sperm contents and sperm motility patterns were significantly altered in all the metal-treated groups, suggesting the direct/indirect spermotoxic effects of lead and cadmium. The inhibitory effects of lead, cadmium and combined exposure on testicular steroidogenesis machinery, along with the male accessory gland functions, are indicative of multiple targets of lead and cadmium to disrupt male reproductive functions.


Subject(s)
Acetates/toxicity , Antioxidants/metabolism , Cadmium/toxicity , Epididymis/metabolism , Lead/toxicity , Reproduction/drug effects , Spermatozoa/drug effects , Testis/drug effects , 17-Hydroxysteroid Dehydrogenases/drug effects , Animals , Ascorbic Acid/metabolism , Catalase/metabolism , Cell Survival/drug effects , Epididymis/drug effects , Female , Lactation/drug effects , Lipid Peroxidation/drug effects , Male , Organometallic Compounds/toxicity , Pregnancy , Rats , Sperm Count , Sperm Motility/drug effects , Steroids/biosynthesis , Superoxide Dismutase/metabolism , Testis/metabolism , Testosterone/blood
10.
J Biochem Mol Toxicol ; 25(1): 15-25, 2011.
Article in English | MEDLINE | ID: mdl-20957662

ABSTRACT

Exposure to toxic metals including cadmium has become an increasingly recognized source of illness worldwide. Cadmium (Cd(2+) ) is one of the environmental pollutants affecting various tissues and organs including testis. The protective effect of lipoic acid and selenium on Cd(2+) -induced testicular damage was investigated. Accordingly, male Wistar rats were allocated into four groups (n = 8; each). Gp I: (control), whereas the other 3 groups received CdCl(2) (2 mg/kg, i.p. for 28 days) alone or in combination with either (i) lipoic acid (35 mg/kg, p.o) or (ii) selenium (0.35 mg/kg, p.o) throughout the experiment. Serum testosterone, luteinizing hormone and follicle-stimulating hormone levels significantly decreased in the Cd(2+) -exposed rats. The activities of testicular key androgenic enzymes, 3ß-hydroxysteroid dehydrogenase and 17 ß-HSD significantly decreased in Cd(2) exposed rats compared to the control counterparts. In addition, the activities of testicular marker enzymes were significantly altered in cadmium-treated animals. Significant reductions in body and testicular weight as well as antioxidant status were also observed in Cd(2+) -exposed rats. Moreover, some testicular metal levels were altered. Lipoic acid and selenium significantly increased serum testosterone level and restored testicular activity of 3ß-HSD and 17 ß-HSD and were effective in modulation of most of the measured biochemical parameters. The biochemical parameters were further confirmed with histopathological findings. In conclusion, the present study demonstrated the beneficial influences of lipoic acid and selenium in reducing harmful effects of Cd(2+) in rats' testes.


Subject(s)
Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/drug effects , Selenium/pharmacology , Testicular Diseases/drug therapy , Testis/drug effects , Testosterone/metabolism , Thioctic Acid/pharmacology , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Body Weight/drug effects , Cadmium , Follicle Stimulating Hormone/blood , Lipid Peroxidation/drug effects , Luteinizing Hormone/metabolism , Luteinizing Hormone/pharmacology , Male , Metals, Heavy/metabolism , Organ Size/drug effects , Rats , Rats, Wistar , Testicular Diseases/chemically induced , Testicular Diseases/metabolism , Testis/metabolism
11.
Asian J Androl ; 12(4): 519-26, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20453869

ABSTRACT

The objective of the present study was to investigate the effects of genistein and equol on 3beta-hydroxysteroid dehydrogenase (3beta-HSD) and 17beta-hydroxysteroid dehydrogenase 3 (17beta-HSD3) in human and rat testis microsomes. These enzymes (3beta-HSD and 17beta-HSD3), along with two others (cytochrome P450 side-chain cleavage enzyme and cytochrome P450 17alpha-hydroxylase/17-20 lyase), catalyze the reactions that convert the steroid cholesterol into the sex hormone testosterone. Genistein inhibited 3beta-HSD activity (0.2 micromol L(-1) pregnenolone) with half-maximal inhibition or a half-maximal inhibitory concentration (IC(50)) of 87 +/- 15 (human) and 636 +/- 155 nmol L(-1) (rat). Genistein's mode of action on 3beta-HSD activity was competitive for the substrate pregnenolonrge and noncompetitive for the cofactor NAD(+). There was no difference in genistein's potency of 3beta-HSD inhibition between intact rat Leydig cells and testis microsomes. In contrast to its potent inhibition of 3beta-HSD, genistein had lesser effects on human and rat 17beta-HSD3 (0.1 micromol L(-1) androstenedione), with an IC(50) >or= 100 micromol L(-1). On the other hand, equol only inhibited human 3beta-HSD by 42%, and had no effect on 3beta-HSD and 17beta-HSD3 in rat tissues. These observations imply that the ability of soy isoflavones to regulate androgen biosynthesis in Leydig cells is due in part to action on Leydig cell 3beta-HSD activity. Given the increasing intake of soy-based food products and their potential effect on blood androgen levels, these findings are greatly relevant to public health.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Genistein/pharmacology , Isoflavones/pharmacology , Testis/drug effects , 17-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/drug effects , Animals , Equol , Humans , Kinetics , Leydig Cells/enzymology , Male , Microsomes/drug effects , Microsomes/enzymology , Rats , Rats, Sprague-Dawley , Testis/enzymology
12.
Oncol Rep ; 23(4): 1173-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20204307

ABSTRACT

Melatonin interacts with estradiol at the estrogen receptor level in different kinds of neoplasias and also regulates the expression and the activity of some enzymes involved in the biosynthesis of estrogens in peripheral tissues. Glioma cells express estrogen receptors and have the ability to synthesize estrogens locally. Since melatonin inhibits the growth of C6 cells, and this indoleamine has been demonstrated to be capable of decreasing aromatase expression and activity in these cells, the aim of the present study was to analyze whether the regulation of the sulfatase, the enzyme that catalyzes the rate-limiting step in the conversion of estrogen sulfates to estrogens, and 17beta-hydroxysteroid dehydrogenase, the enzyme which converts the relatively inactive estrone to the most potent 17beta-estradiol, could be involved in the inhibition of glioma cell growth by melatonin. We found that melatonin decreases the growth of C6 glioma cells and reduces the sulfatase and 17beta-hydroxysteroid dehydrogenase activity. Finally, we demonstrated that melatonin downregulates sulfatase and 17beta-hydroxysteroid dehydrogenase mRNA steady state levels in these glioma cells. By analogy to the implications of these enzymes in other forms of estrogen-sensitive tumors, it is conceivable that their modulation by melatonin may play a role in the growth of glioblastomas.


Subject(s)
17-Hydroxysteroid Dehydrogenases/drug effects , Antioxidants/pharmacology , Glioma/enzymology , Melatonin/pharmacology , Sulfatases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Cell Line, Tumor , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sulfatases/metabolism
13.
Toxicol Appl Pharmacol ; 245(1): 124-33, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20214916

ABSTRACT

In this study, we have identified several ovarian steroids in Ciona with high similarity to vertebrate steroids and showed that cholesterol, corticosterone, dehydroepiandrosterone, estrone, estradiol-17beta, testosterone, pregnenolone, progesterone, have identical molecular spectra with vertebrate steroids. In addition, we have studied the effects of an endocrine disruptor (tributyltin: TBT) on these sex hormones and their precursors, ovarian morphology, and gene expression of some key enzymes in steroidogenic pathway in the ovary of Ciona. Ovarian specimens were cultured in vitro using different concentrations of TBT (10(-5), 10(-4) and 10(-3)M). Ethanol was used as solvent control. Gene expression analysis was performed for adrenodoxin (ADREN) and adrenodoxin reductase (ADOX) (mediators of acute steroidogenesis) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD). These transcripts were detected and measured by quantitative (real-time) polymerase chain reaction (qPCR). Sex steroids and their precursors were identified and quantified by a gas chromatography-mass spectroscopy (GC-MS) method. Exposure of Ciona ovaries to TBT produced modulations (either increased or decreased) of sterols and sex steroid levels, whereas no significant differences in ADREN, ADOX or 17beta-HSD mRNA expression patterns were observed. Histological analysis shows that TBT produced several modifications on Ciona ovarian morphology that includes irregular outline of nuclear membrane, less compacted cytoplasm, in addition to test and granulosa cells that were detached from the oocyte membrane. Given that the ascidians represent very simple experimental models for the study of endocrine disruption by environmental contaminants, our findings provide excellent models for multiple identification and quantification of sex steroid and their precursors in biological samples exposed to endocrine-disrupting chemicals and for direct extrapolation of such effects across taxonomic groups and phyla. In addition, these results suggest that Cionaintestinalis may be a suitable species for molecular ecotoxicological studies and biomarker model for endocrine-disrupting effects in marine invertebrates.


Subject(s)
Ciona intestinalis/drug effects , Endocrine Disruptors/toxicity , Gonadal Steroid Hormones/metabolism , Ovary/drug effects , Trialkyltin Compounds/toxicity , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Adrenodoxin/drug effects , Adrenodoxin/genetics , Adrenodoxin/metabolism , Animals , Cholesterol/analysis , Ciona intestinalis/chemistry , Ciona intestinalis/physiology , Corticosterone/analysis , Dehydroepiandrosterone/analysis , Estradiol/analysis , Estrone/analysis , Female , Ferredoxin-NADP Reductase/drug effects , Ferredoxin-NADP Reductase/genetics , Ferredoxin-NADP Reductase/metabolism , Gas Chromatography-Mass Spectrometry , Gene Expression/drug effects , Gonadal Steroid Hormones/genetics , Ovary/anatomy & histology , Ovary/physiology , Pregnenolone/analysis , Progesterone/analysis , Testosterone/analysis
14.
J Physiol Pharmacol ; 60(1): 181-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19439821

ABSTRACT

Overweight male rats received oral oleoyl-estrone (OE) for 10 days, and were compared with controls. The expression of 17beta-hydroxysteroid dehydrogenase (17betaHSDH) isoenzymes, and other proteins related to sex hormone metabolism, were analyzed in testicle, liver, adrenals and two white adipose sites: subcutaneous inguinal and epididymal pads using a semiquantitative RT-PCR method. Androstenedione, testosterone, estrone and estradiol levels were measured by HPLC-MS/MS. Isoenzyme expressions were grouped according to their main physiological function (oxidative or reductive) and preferred substrate (androgen or estrogen). As expected, testicle was the main site for synthesis of testosterone and estradiol, and the liver the main organ oxidizing them to androstenedione and estrone. Overall oxidative capacity was 6.5-fold higher than the reductive, and estradiol synthesis and oxidation potential were higher than for testosterone. OE decreased serum androgens, and increased estrone, but not estradiol. This was due to decreased testicle ability to produce testosterone, because of smaller size and decreased 17betaHSDH3 expression, but also to lower availability of precursors. High estrone availability (from OE hydrolysis) does not translate into higher estradiol because of decreased testicle reductive 17betaHSDH expression and decreased aromatase. In consequence, we can assume that OE effects on androgens, and the hypothalamic-pituitary-gonadal axis are limited to testicles.


Subject(s)
17-Hydroxysteroid Dehydrogenases/drug effects , Anti-Obesity Agents/pharmacology , Estrone/analogs & derivatives , Oleic Acids/pharmacology , Testosterone/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Chromatography, High Pressure Liquid , Estradiol/metabolism , Estrone/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes , Male , Overweight , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Testis/metabolism
15.
Curr Cancer Drug Targets ; 8(8): 691-702, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19075592

ABSTRACT

Melatonin exerts oncostatic effects on different kinds of tumors, especially on hormone-dependent breast cancer. The general conclusion is that melatonin, in vivo, reduces the incidence and growth of chemically-induced mammary tumors in rodents, and, in vitro, inhibits the proliferation and invasiveness of human breast cancer cells. Both studies support the hypothesis that melatonin inhibits the growth of breast cancer by interacting with estrogen-signaling pathways through three different mechanisms: (a) the indirect neuroendocrine mechanism which includes the melatonin down-regulation of the hypothalamic-pituitary-reproductive axis and the consequent reduction of circulating levels of gonadal estrogens, (b) direct melatonin actions at tumor cell level by interacting with the activation of the estrogen receptor, thus behaving as a selective estrogen receptor modulator (SERM), and (c) the regulation of the enzymes involved in the biosynthesis of estrogens in peripheral tissues, thus behaving as a selective estrogen enzyme modulator (SEEM). As melatonin reduces the activity and expression of aromatase, sulfatase and 17beta-hydroxysteroid dehydrogenase and increases the activity and expression of estrogen sulfotransferase, it may protect mammary tissue from excessive estrogenic effects. Thus, a single molecule has both SERM and SEEM properties, one of the main objectives desired for the breast antitumoral drugs. Since the inhibition of enzymes involved in the biosynthesis of estrogens is currently one of the first therapeutic strategies used against the growth of breast cancer, melatonin modulation of different enzymes involved in the synthesis of steroid hormones makes, collectively, this indolamine an interesting anticancer drug in the prevention and treatment of estrogen-dependent mammary tumors.


Subject(s)
Breast Neoplasms/enzymology , Melatonin/pharmacology , Neoplasms, Hormone-Dependent/enzymology , Selective Estrogen Receptor Modulators/pharmacology , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Aromatase/drug effects , Aromatase/metabolism , Breast Neoplasms/physiopathology , Estrogens/physiology , Humans , Melatonin/physiology , Neoplasms, Hormone-Dependent/physiopathology , Sulfatases/drug effects , Sulfatases/metabolism
16.
Arch Toxicol ; 82(11): 851-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18449530

ABSTRACT

We recently showed that prenatal exposure to diesel exhaust (DE) disrupts spermatogenesis in mouse offspring. This study was undertaken to determine whether filtered DE in which 99.97% of diesel exhaust particles >0.3 microm in diameter were removed affects spermatogenesis in growing mice. After prenatal exposure to filtered DE for 2-16 days postcoitum, we examined daily sperm production (DSP), testicular histology, serum testosterone levels and mRNA expression of hormone synthesis process-related factors. In the filtered DE exposed group, DSP was markedly reduced at 12 weeks compared with the control group; clean air exposed group. Histological examination showed multinucleated giant cells and partial vacuolation in the seminiferous tubules of the exposed group. Testosterone was elevated significantly at 5 weeks. Moreover, luteinizing hormone receptor mRNA at 5 and 12 weeks, 17alpha-hydroxylase/C17-20-lyase and 17beta-hydroxysteroid dehydrogenase mRNAs at 12 weeks were significantly elevated. These results suggest that filtered DE retains its toxic effects on the male reproductive system following prenatal exposure.


Subject(s)
Prenatal Exposure Delayed Effects , Spermatogenesis/drug effects , Testis/drug effects , Vehicle Emissions/toxicity , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Female , Gene Expression Regulation/drug effects , Giant Cells/drug effects , Giant Cells/metabolism , Male , Mice , Mice, Inbred ICR , Particle Size , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, LH/drug effects , Receptors, LH/metabolism , Seminiferous Tubules/drug effects , Sperm Count , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism , Testis/metabolism , Testosterone/blood , Time Factors , Vacuoles/drug effects , Vacuoles/metabolism
17.
Arch Toxicol ; 82(11): 833-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18401575

ABSTRACT

Methoxychlor, an organochlorine pesticide, has been reported to induce reproductive abnormalities in male reproductive tract. To get more insight into the mechanism(s) of gonadal toxicity provoked by methoxychlor, we investigated whether treatment with methoxychlor at low observed adverse effect level (LOAEL) would alter the activities of steroidogenic enzymes such as Delta(5)3beta-hydroxysteroid dehydrogenase (3beta-HSD) and Delta(5)17beta-hydroxysteroid dehydrogenase (17beta-HSD), the expression levels of steroidogenic acute regulatory (StAR) protein and androgen binding protein (ABP) in the testis of adult male rats. The experimental rats were exposed to a single dose of methoxychlor (50 mg/kg body weight) orally. The rats were killed at 0, 3, 6, 12, 24 and 72 h following treatment using anesthetic ether and testes were collected, processed and used to measure the activities of 3beta-HSD, 17beta-HSD, levels of hydrogen peroxide produced and the expression levels of StAR protein, and ABP. Methoxychlor administration resulted in a sequential reduction in the expression of StAR protein and activities of 3beta-HSD, 17beta-HSD with concomitant increase in the levels of hydrogen peroxide in the testis. These changes were significant between 6-12 h following treatment. The levels of ABP declined at 6-12 h following exposure to methoxychlor. The present study demonstrates transient effect of methoxychlor at LOAEL on testicular steroidogenesis and the possible role of hydrogen peroxide in mediating these effects.


Subject(s)
Gene Expression Regulation/drug effects , Insecticides/toxicity , Methoxychlor/toxicity , Testis/drug effects , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/metabolism , Administration, Oral , Androgen-Binding Protein/drug effects , Androgen-Binding Protein/metabolism , Animals , Hydrogen Peroxide/metabolism , Insecticides/administration & dosage , Male , Methoxychlor/administration & dosage , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Rats , Rats, Wistar , Testis/metabolism , Time Factors
18.
Int J Cancer ; 122(9): 1931-40, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18183589

ABSTRACT

Oestradiol (E2) stimulates the growth of hormone-dependent breast cancer. 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyse the pre-receptor activation/inactivation of hormones and other substrates. 17beta-HSD1 converts oestrone (E1) to active E2, but it has recently been suggested that another 17beta-HSD, 17beta-HSD12, may be the major enzyme that catalyses this reaction in women. Here we demonstrate that it is 17beta-HSD1 which is important for E2 production and report the inhibition of E1-stimulated breast tumor growth by STX1040, a non-oestrogenic selective inhibitor of 17beta-HSD1, using a novel murine model. 17beta-HSD1 and 17beta-HSD12 mRNA and protein expression, and E2 production, were assayed in wild type breast cancer cell lines and in cells after siRNA and cDNA transfection. Although 17beta-HSD12 was highly expressed in breast cancer cell lines, only 17beta-HSD1 efficiently catalysed E2 formation. The effect of STX1040 on the proliferation of E1-stimulated T47D breast cancer cells was determined in vitro and in vivo. Cells inoculated into ovariectomised nude mice were stimulated using 0.05 or 0.1 microg E1 (s.c.) daily, and on day 35 the mice were dosed additionally with 20 mg/kg STX1040 s.c. daily for 28 days. STX1040 inhibited E1-stimulated proliferation of T47D cells in vitro and significantly decreased tumor volumes and plasma E2 levels in vivo. In conclusion, a model was developed to study the inhibition of the major oestrogenic 17beta-HSD, 17beta-HSD1, in breast cancer. Both E2 production and tumor growth were inhibited by STX1040, suggesting that 17beta-HSD1 inhibitors such as STX1040 may provide a novel treatment for hormone-dependent breast cancer.


Subject(s)
17-Hydroxysteroid Dehydrogenases/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Estrogens/blood , Estrone/analogs & derivatives , Mammary Neoplasms, Experimental/enzymology , Neoplasms, Hormone-Dependent/enzymology , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Cell Line, Tumor , Chromatography, High Pressure Liquid , DNA, Complementary/metabolism , Estradiol/blood , Estrogens/metabolism , Estrone/blood , Estrone/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/blood , Neoplasms, Hormone-Dependent/drug therapy , Ovariectomy , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Nepal Med Coll J ; 8(2): 101-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-17017399

ABSTRACT

Adult male rats received daily injections (sc) of estradiol-17 beta (50 microg/100 g body wt per day) for 7 days. When they were sacrificed 14 days after the last injection, serum levels of gonadotropins and testosterone and weights of accessory sex organs were decreased significantly, testicular 17-hydroxysteroid dehydroganase activity was suppressed and spermatogenesis was inhibited in 5.0% casein-fed estrogen-treated rats. Feeding of 20.0% casein diet to estrogen-treated rats resulted in increased serum concentration of gonadotropins and testosterone. LH and testosterone appeared to be normal in 20.0% casein-fed estrogen-treated rats while serum FSH levels remained subnormal. The estrogen-treated rats fed on 20.0% casein diet showed decreased spermatogenesis in comparison with control rats fed on 20.0% casein diet. Together, these results indicate that high casein diet stimulates synthesis of testicular testosterone and increases serum LH levels more than FSH in estrogen-treated rats. It is concluded that estrogen in the presence of high milk protein diet may be considered to be a suitable steroid hormone in the development of a male contraceptive.


Subject(s)
Contraceptive Agents/pharmacology , Estradiol/pharmacology , Milk Proteins , Spermatogenesis/drug effects , 17-Hydroxysteroid Dehydrogenases/drug effects , Animals , Contraceptive Agents/adverse effects , Estradiol/adverse effects , Follicle Stimulating Hormone , Male , Rats , Rats, Wistar
20.
Biol Reprod ; 75(4): 605-14, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16807381

ABSTRACT

The opposing actions of estrogen and progesterone during the menstrual cycle regulate the cyclical and predictable endometrial proliferation and differentiation that is required for implantation. Progesterone indirectly stimulates the expression of 17beta hydroxysteroid dehydrogenase type 2 (HSD17B2), which catalyzes the conversion of biologically potent estradiol to weakly estrogenic estrone in the endometrial epithelium. We previously demonstrated upregulation of the HSD17B2 gene in human endometrial epithelial cells by factors secreted from endometrial stromal cells in response to progesterone. We investigated the underlying mechanism by which these stroma-derived, progesterone-induced paracrine factors stimulate HSD17B2 expression. Here, we show that transcription factors SP1 and SP3 interact with specific motifs in HSD17B2 promoter to upregulate enzyme expression in human endometrial epithelial cell lines. Conditioned medium (CM) from progestin-treated stromal cells increased levels of SP1 and SP3 in endometrial epithelial cells and induced HSD17B2 mRNA expression. Mithramycin A, an inhibitor of SP1-DNA interaction, reduced epithelial HSD17B2 promoter activity in a dose-dependent manner. Serial deletion and site-directed mutants of the HSD17B2 promoter demonstrated that two overlapping SP1 motifs (nt -82/-65) are essential for induction of promoter activity by CM or overexpression of SP1/SP3. CM markedly enhanced, whereas anti-SP1/SP3 antibodies inhibited, binding of nuclear proteins to this region of the HSD17B2 promoter. In vivo, we demonstrated a significant spatiotemporal association between epithelial SP1/SP3 and HSD17B2 levels in human endometrial biopsies. Taken together, these data suggest that HSD17B2 expression in endometrial epithelial cells, and, therefore, estrogen inactivation, is regulated by SP1 and SP3, which are downstream targets of progesterone-dependent paracrine signals originating from endometrial stromal cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Endometrium/metabolism , Progesterone/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Adult , Base Sequence , Binding Sites , Cells, Cultured , Epithelial Cells/metabolism , Estradiol Dehydrogenases , Female , GC Rich Sequence , Humans , Middle Aged , Molecular Sequence Data , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Pregnancy , Promoter Regions, Genetic , Sequence Deletion , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...