Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506
Filter
1.
Reprod Biol Endocrinol ; 19(1): 74, 2021 May 17.
Article in English | MEDLINE | ID: mdl-34001150

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder with various manifestations and complex etiology. Follicular fluid (FF) serves as the complex microenvironment for follicular development. However, the correlation between the concentration of steroid in FF and the pathogenesis of PCOS is still unclear. METHODS: Twenty steroid levels in FF from ten patients with PCOS and ten women with male-factor infertility undergoing in vitro fertilization were tested by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in order to explore their possibly correlation with PCOS. Meanwhile, the mRNA levels of core enzymes in steroid synthesis pathway from exosomes of FF were also detected by qPCR. RESULTS: The estriol (p < 0.01), estradiol (p < 0.05) and prenenolone (p < 0.01) levels in FF of PCOS group were significantly increased, compared to the normal group, and the progesterone levels (p < 0.05) were decreased in PCOS group. Increased mRNA levels of CYP11A, CYP19A and HSD17B2 of exosomes were accompanied by the hormonal changes in FF. Correlation analysis showed that mRNA levels of CYP11A and HSD17B2 were negatively correlated with percent of top-quality embryos and rate of embryos develop to blastocyst. CONCLUSION: Our results suggest that increased levels of estrogen and pregnenolone in follicular fluid may affect follicle development in PCOS patients, and the mechanism is partially related to HSD17B1, CYP19A1 and CYP11A1 expression change in FF exosomes.


Subject(s)
Exosomes/metabolism , Follicular Fluid/chemistry , Ovulation Induction , Polycystic Ovary Syndrome/metabolism , Steroids/analysis , Adult , Aromatase/biosynthesis , Aromatase/genetics , Blastocyst/cytology , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/genetics , Chromatography, Liquid , Embryonic Development , Estradiol/analysis , Estradiol Dehydrogenases/biosynthesis , Estradiol Dehydrogenases/genetics , Estriol/analysis , Exosomes/ultrastructure , Female , Humans , Nanoparticles , Oocyte Retrieval , Ovulation Induction/methods , Pregnenolone/analysis , Progesterone/analysis , RNA, Messenger/biosynthesis , Tandem Mass Spectrometry
2.
Endocrinology ; 162(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-33831176

ABSTRACT

Elucidating the global molecular changes that occur during aromatase inhibitor (AI)- or 17α-methyltestosterone (MT)-induced masculinization and estradiol-17ß (E2)-induced feminization is critical to understanding the roles that endocrine and genetic factors play in regulating the process of sex differentiation in fish. Here, fugu larvae were treated with AI (letrozole), MT, or E2 from 25 to 80 days after hatching (dah), and gonadal transcriptomic analysis at 80 dah was performed. The expression of dmrt1, gsdf, foxl2, and other key genes (star, hsd3b1, cyp11c1, cyp19a1a, etc.) involved in the steroid hormone biosynthesis pathway were found be altered. The expression of dmrt1, gsdf, cyp19a1a, and foxl2 was further verified by quantitative polymerase chain reaction. In the control group, the expression of dmrt1 and gsdf was significantly higher in XY larvae than in XX larvae, while the expression of foxl2 and cyp19a1a was significantly higher in XX larvae than in XY larvae (P < .05). AI treatment suppressed the expression of foxl2 and cyp19a1a, and induced the expression of dmrt1 and gsdf in XX larvae. MT treatment suppressed the expression of foxl2, cyp19a1a, dmrt1, and gsdf in XX larvae. E2 treatment suppressed the expression of dmrt1 and gsdf, but did not restore the expression of foxl2 and cyp19a1a in XY larvae. The shared response following AI, MT, and E2 treatment suggested that these genes are essential for sex differentiation. This finding offers some insight into AI or MT-induced masculinization, and E2-induced femininization in fugu.


Subject(s)
Aromatase Inhibitors/pharmacology , Estradiol/pharmacology , Feminization/metabolism , Gene Expression Profiling , Gene Expression Regulation , Methyltestosterone/pharmacology , Takifugu/metabolism , Animals , Aromatase/biosynthesis , Female , Forkhead Box Protein L2/biosynthesis , Gonads/metabolism , Letrozole/pharmacology , Male , Polymerase Chain Reaction , RNA-Seq , Sex Differentiation/drug effects , Transcription Factors/biosynthesis , Transcriptome/drug effects
3.
J Comp Neurol ; 528(15): 2499-2522, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32190905

ABSTRACT

Estrogen synthesis and signaling in the brains of vertebrates has pleotropic effects ranging from neurogenesis to modulation of behaviors. The majority of studies on brain-derived estrogens focus on males, but estrogenic signaling in females likely plays important roles in regulation of reproductive cycling and social behaviors. We used females of the mouth brooding African cichlid fish, Astatotilapia burtoni, to test for reproductive state-dependent changes in estrogenic signaling capacity within microdissected brain nuclei that are important for social behaviors. Expression levels of the rate-limiting enzyme aromatase, but not estrogen receptors, measured by qPCR changes across the reproductive cycle. Gravid females that are close to spawning had higher aromatase levels in all brain regions compared to females with lower reproductive potential. This brain aromatase expression was positively correlated with circulating estradiol levels and ovarian readiness. Using chromogenic in situ hybridization we localized aromatase-expressing cells to ependymal regions bordering the ventricles from the forebrain to the hindbrain, and observed more abundant staining in gravid compared to mouth brooding females in most regions. Staining was most prominent in subpallial telencephalic regions, and diencephalic regions of the preoptic area, thalamus, and hypothalamus, but was also observed in sensory and sensorimotor areas of the midbrain and hindbrain. Aromatase expression was observed in radial glial cells, revealed by co-localization with the glial marker GFAP and absence of co-localization with the neuronal marker HuC/D. Collectively these results support the idea that brain-derived estradiol in females may serve important functions in reproductive state-dependent physiological and behavioral processes across vertebrates.


Subject(s)
Aromatase/biosynthesis , Brain/metabolism , Cichlids/metabolism , Genitalia, Female/metabolism , Receptors, Estrogen/biosynthesis , Reproduction/physiology , Animals , Aromatase/genetics , Cichlids/genetics , Female , Gene Expression , Male , Receptors, Estrogen/genetics
4.
Ecotoxicol Environ Saf ; 193: 110324, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32088548

ABSTRACT

This study assessed the transcription levels of estrogen-responsive genes, such as vitellogenins (Vtg1 and Vtg2), choriogenins (ChgL, ChgH, and ChgHm), cytochrome P450 aromatase (CYP19a1b), and ER subtypes (ERα, ERß1, and ERß2), in 7 days-post-fertilization (dpf) embryos and 9 and 12 dpf larvae of medaka (Oryzias latipes) exposed to estrogenic endocrine-disrupting chemicals (EDCs). The <5 h-post-fertilization embryos were exposed to EDCs such as 17ß-estradiol (E2), p-n-nonylphenol (NP), and bisphenol A (BPA). In E2 (0.10-222 nM)-treated 7 dpf embryos and 9 or 12 dpf larvae, ChgL, ChgH, and ChgHm expression was up-regulated in a concentration-dependent manner. By contrast, interestingly, Vtg1 and Vtg2 expression was not induced in E2-treated 7 dpf embryos but was significantly induced in 9 and 12 dpf larvae, suggesting a developmental-stage-specific regulatory mechanism underlying Vtg expression. The maximum concentrations of NP (0.09-1.5 µM) and BPA (1.8-30 µM) up-regulated Chg expression in 9 or 12 dpf larvae, and the relative estrogenic potencies (REPs) of E2, NP, and BPA were 1, 2.1 × 10-4, and 1.0 × 10-5, respectively. Chg messenger RNA (mRNA) in medaka embryos and larvae can be used as a sensitive biomarker for screening potential estrogenic EDCs. Our assay system using embryos and larvae can be used as an in vivo alternative model because independent feeding stages (e.g., embryonic and early larval stages) are suitable alternatives.


Subject(s)
Egg Proteins/biosynthesis , Endocrine Disruptors/toxicity , Estrogens/toxicity , Oryzias/embryology , Oryzias/growth & development , Animals , Aromatase/biosynthesis , Aromatase/genetics , Benzhydryl Compounds/toxicity , Egg Proteins/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Estradiol/toxicity , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Larva/drug effects , Larva/genetics , Larva/metabolism , Male , Models, Animal , Oryzias/genetics , Phenols/toxicity , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Vitellogenins/biosynthesis , Vitellogenins/genetics
5.
J Neuroendocrinol ; 31(12): e12816, 2019 12.
Article in English | MEDLINE | ID: mdl-31758603

ABSTRACT

Phthalates are found in different plastic materials, such as packaging, toys and medical devices. Some of these compounds are endocrine disruptors, comprising substances that are able to induce multiple hormonal disturbances and downstream developmental effects, including the disruption of androgen-dependent differentiation of the male reproductive tract and changes in pathways that regulate hormone-dependent behaviours. In a previous study, metabolites of diisopentyl phthalate (DiPeP), a potent anti-androgenic phthalate, were found in the urine of Brazilian pregnant women. Therefore, the present study aimed to evaluate the effects of DiPeP exposure during critical developmental periods on behaviours controlled by sex hormones in rats. Pregnant Wistar rats were treated with DiPeP (1, 10 or 100 mg kg day-1 ) or canola oil by oral gavage between gestational day 10 and post-natal day (PND) 21. Male offspring were tested in a behavioural battery, including the elevated plus maze task, play behaviour, partner preference and sexual behaviour. After the behavioural tests, the hypothalamus and pituitary of these animals were removed on PND 60-65 and PND 145-160 to quantify gene expression for aromatase, androgen receptor (Ar) and oestrogen receptors α (Esr1) and ß (Esr2). Male rats exposed to 1 and 10 mg kg day-1 DiPeP displayed no preference for the female stimulus rat in the partner preference test and 1 mg kg day-1 DiPeP rats also showed a significant increase in mount and penetration latencies when mated with receptive females. A decrease in pituitary Esr1 expression was observed in all DiPeP treated groups regardless of age. A reduction in hypothalamic Esr1 expression in rats exposed to 10 mg kg day-1 DiPeP was also observed. No significant changes were found with respect to Ar, Esr2 and aromatase expression in the hypothalamus. These results suggest that DiPeP exposure during critical windows of development in rats may induce changes in behaviours related to mating and the sexual motivation of males.


Subject(s)
Aromatase/biosynthesis , Behavior, Animal/drug effects , Behavior, Animal/physiology , Prenatal Exposure Delayed Effects/physiopathology , Receptors, Androgen/biosynthesis , Receptors, Estrogen/biosynthesis , Animals , Dose-Response Relationship, Drug , Female , Hypothalamus/metabolism , Lactation , Male , Pituitary Gland/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats
6.
Mol Reprod Dev ; 86(12): 1901-1908, 2019 12.
Article in English | MEDLINE | ID: mdl-31713287

ABSTRACT

Follicle growth and granulosa cell health are dependent on the secretion of estradiol from granulosa cells. Estradiol is synthesized from androgen precursor by cytochrome P450 aromatase (CYP19A1), and in cattle CYP19A1 messenger RNA has a short half-life but a long (3.5 kb) 3'-untranslated region (3'UTR), suggesting that posttranscriptional regulation may be important for control of enzyme activity. We tested this hypothesis by inserting the CYP19A1 3'UTR and fragments thereof into a reporter vector between the end of the luciferase coding region and the polyadenylation signal. The full-length aromatase 3'UTR suppressed luciferase activity to 10% of control levels, and smaller fragments showed that this inhibitory activity lies between +926 and +1134 of the 3'UTR. Protein-RNA cross-linking experiments revealed that these 3'UTR fragments formed an RNA-protein complex of approximately 70 kDa that was present in granulosa cells but not in corpus luteum, lung, liver, kidney, pancreas, or bladder extracts. The RNA-binding activity was specific to the 3'UTR, as shown by competition experiments with unlabeled RNA, and was present only in 3'UTR constructs that inhibited luciferase activity. These data suggest that posttranscriptional regulation is an important component of the control of CYP19A1 expression and involves protein binding to a specific sequence in the 3'UTR.


Subject(s)
3' Untranslated Regions , Aromatase/biosynthesis , Granulosa Cells/metabolism , Multiprotein Complexes/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Animals , Cattle , Female , Granulosa Cells/cytology
7.
Cancer Res ; 79(20): 5342-5354, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31481501

ABSTRACT

Obese women have higher risk of bearing breast tumors that are highly aggressive and resistant to therapies. Tumor-promoting effects of obesity occur locally via adipose inflammation and related alterations to the extracellular matrix (ECM) as well as systemically via circulating metabolic mediators (e.g., free fatty acids, FFA) associated with excess adiposity and implicated in toll-like receptor-mediated activation of macrophages-key cellular players in obesity-related cancer progression. Although the contribution of macrophages to proneoplastic effects of obesity is well documented, the role of ECM components and their enzymatic degradation is less appreciated. We show that heparanase, the sole mammalian endoglucuronidase that cleaves heparan sulfate in ECM, is preferentially expressed in clinical/experimental obesity-associated breast tumors. Heparanase deficiency abolished obesity-accelerated tumor progression in vivo. Heparanase orchestrated a complex molecular program that occurred concurrently in adipose and tumor tissue and sustained the cancer-promoting action of obesity. Heparanase was required for adipose tissue macrophages to produce inflammatory mediators responsible for local induction of aromatase, a rate-limiting enzyme in estrogen biosynthesis. Estrogen upregulated heparanase in hormone-responsive breast tumors. In subsequent stages, elevated levels of heparanase induced acquisition of procancerous phenotype by tumor-associated macrophages, resulting in activation of tumor-promoting signaling and acceleration of breast tumor growth under obese conditions. As techniques to screen for heparanase expression in tumors become available, these findings provide rational and a mechanistic basis for designing antiheparanase approaches to uncouple obesity and breast cancer in a rapidly growing population of obese patients. SIGNIFICANCE: This study reveals the role of heparanase in promoting obesity-associated breast cancer and provides a mechanistically informed approach to uncouple obesity and breast cancer in a rapidly growing population of obese patients.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma/enzymology , Glucuronidase/physiology , Obesity/complications , Adipose Tissue/metabolism , Animals , Aromatase/biosynthesis , Aromatase/genetics , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Carcinoma/etiology , Carcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Diet, High-Fat/adverse effects , Disease Progression , Estrogens/physiology , Female , Glucuronidase/deficiency , Glucuronidase/genetics , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/pathology , Pancreatic Neoplasms/pathology
8.
Mol Reprod Dev ; 86(8): 1023-1032, 2019 08.
Article in English | MEDLINE | ID: mdl-31179605

ABSTRACT

Long noncoding RNAs (lncRNAs) have been demonstrated to play vital roles in mammalian reproduction. Our previous research revealed that lncRNA Gm2044 is highly expressed in mouse spermatocytes and regulates male germ cell function. The gene annotation database BioGPS shows that Gm2044 is not only highly expressed in testicular tissue but also in ovarian tissue, which suggests that Gm2044 may be involved in female reproductive development. In this study, we confirmed that lncRNA Gm2044 promotes 17ß-estradiol synthesis in mouse pre-antral follicular granulosa cells (mpGCs). Furthermore, bioinformatics methods, western blot, and the luciferase assay proved that Gm2044 functions as a miR-138-5p sponge to inhibit the direct target of miR-138-5p, Nr5a1, which enhances 17ß-estradiol synthesis through cyp19a1 activation. Taken together, our results provide an insight into the mechanistic roles of lncRNA Gm2044 for 17ß-estradiol synthesis by acting as competing-endogenous RNAs to modulate the function of mpGCs. Studying the potential lncRNAs, which regulate estradiol release, will be beneficial for the diagnosis and treatment of steroid hormone-related disease.


Subject(s)
Estradiol/biosynthesis , Granulosa Cells/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Aromatase/biosynthesis , Female , Mice , Mice, Inbred ICR , Steroidogenic Factor 1/biosynthesis
9.
Tissue Cell ; 58: 70-75, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31133248

ABSTRACT

Aromatase plays a central role in ovarian differentiation and development in teleosts. In the present study, we identified a cyp19a1a homologue from the ovary of Schizothorax prenanti and analysed its expression at both the mRNA and protein levels. Cyp19a1a of S. prenanti showed high homology with that of other teleosts, especially S. kozlovi. The ovary and testis were the main sites of cyp19a1a expression in S. prenanti, and cyp19a1a transcript levels peaked in the mid-vitellogenic (MVG)-stage ovary and the mid-spermatogenic (MS)-stage testis. Signals of Cyp19a1a immunopositivity were detected in the spermatocytes and follicular cells of cortical alveolar-stage and MVG oocytes but not in spermatogonia or spermatids. Taken together, these findings indicate that Cyp19a1a may play an important role in oocyte vitellogenesis as well as spermatocyte development in S. prenanti.


Subject(s)
Aromatase/biosynthesis , Cyprinidae/metabolism , Gene Expression Regulation, Enzymologic , Ovary/enzymology , Testis/enzymology , Animals , Aromatase/genetics , Cyprinidae/genetics , Female , Fish Proteins , Male , Oocytes/enzymology , Spermatocytes/enzymology
10.
J Biochem Mol Toxicol ; 33(6): e22317, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30817060

ABSTRACT

This study was aimed to predict bisphenol-A (BPA)-responsive miRNA's using an in silico approach and to study their expression in granulosa cells of animals exposed prenatally to BPA. Pregnant Wistar rats were exposed to BPA through water (25 µg/L, 250 µg/L, and 2.5 mg/L) during gestation. The expression of miRNA-133b, miRNA-378 and miRNA-224 were analyzed in ovarian granulosa cells. BPA affected the postnatal developmental landmarks such as weight of the pups at birth and reduced anogenital distance. BPA exposed animals showed elevated serum estradiol (E2) levels, while follicle-stimulating hormone levels were reduced. The expression of miRNA-224 and aromatase protein levels were found to be increased. This preliminary finding reveals the impact of early life exposure to BPA on the long-term ovarian functions that may be mediated through miRNA-based granulosa cell response. Besides, it is also a compelling indicator for the subclinical response that could have important consequences on female fertility.


Subject(s)
Aromatase/biosynthesis , Benzhydryl Compounds/toxicity , Estradiol/blood , Gene Expression Regulation, Enzymologic/drug effects , Granulosa Cells/metabolism , MicroRNAs/biosynthesis , Phenols/toxicity , Prenatal Exposure Delayed Effects/metabolism , Animals , Female , Granulosa Cells/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar
11.
Histochem Cell Biol ; 151(1): 21-28, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30159783

ABSTRACT

Parietal cells in the gastric mucosa are known not only as cells playing major roles in food digestion but also as cells bearing endocrine function. In addition to their production of gastrin and ghrelin, it has been recently revealed that these cells are also involved in the synthesis and secretion of estrogens with their expression of aromatase in experimental animals. Although aromatase activity has been detected in human gastric cancer cells and related cell lines, much less study has been done to ascertain the expression of the enzymatic activity in normal gastric mucosa. It has not been established which cell type is responsible for estrogen production in human gastric glands consisting of epithelial cells of several types. The aim of this study is to define the expression of aromatase by parietal cells in human gastric glands using immunohistochemical techniques. We retrieved formalin-fixed paraffin embedded materials of gastric biopsies from 16 patients (nine men, seven women). Colocalization of aromatase and H+/K+-ATPase ß-subunit indicated that positive cells are parietal cells, but not chief cells and mucous cells. Furthermore, immunoreactivity of aromatase was detected within gastric glands irrespective of age or sex. These results suggest that human parietal cells synthesize estrogens within gastric mucosa and subsequently secrete them to the portal vein via gastric vein, as they do in rats. These estrogens might influence liver functions in humans. The estrogenic effects related to liver dysfunction might also be attributed to them.


Subject(s)
Aromatase/analysis , Aromatase/biosynthesis , Gastric Mucosa/enzymology , Parietal Cells, Gastric/enzymology , Aromatase/metabolism , Biopsy , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/pathology
12.
Sci Rep ; 8(1): 8722, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880879

ABSTRACT

Reelin plays an important role in cerebral cortex development and synaptogenesis. In the hippocampus, the neurosteroid estrogen affects reelin expression. In this study we tested a potential crosstalk between estradiol and reelin, thus the possibility of a reelin-induced activation of the estradiol synthesizing enzyme aromatase. As a model system, we used ovaries, which express reelin and are a major source of estradiol. We found that in wild-type mice, reelin and aromatase are expressed in granulosa cells of growing follicles. The expression of reelin varies with the estrus cycle and is highest shortly before ovulation, when estradiol serum levels are at their maximum. In ovaries of reelin-deficient reeler mice, aromatase mRNA and protein are significantly reduced, as evidenced by real-time PCR, western blot analysis, and quantitative immunohistochemistry in granulosa cells of preovulatory follicles. In line with reduced estradiol synthesis, ovarian estrus cycle length is prolonged in reeler mice. Most importantly, treating cultured granulosa cells with recombinant reelin results in significant upregulation of aromatase mRNA and protein and increased secretion of estradiol into the supernatant. Our data provide evidence of a local increase of aromatase expression by reelin. Regarding reproduction, this crosstalk may contribute to follicular stability and counteract luteinization in ovaries.


Subject(s)
Aromatase/biosynthesis , Cell Adhesion Molecules, Neuronal/biosynthesis , Estrous Cycle/physiology , Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Granulosa Cells/metabolism , Luteinization/physiology , Nerve Tissue Proteins/biosynthesis , Serine Endopeptidases/biosynthesis , Animals , Female , Granulosa Cells/cytology , Mice , Rats, Wistar , Reelin Protein
13.
Environ Pollut ; 240: 615-622, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29772511

ABSTRACT

Waterborne microcystin-LR (MC-LR) has been reported to disrupt sex hormones, while its estrogenic potency remains controversial. We hypothesized that MC-LR could induce estrogenic effects via disrupting sex hormone synthesis, and verified this hypothesis by in vitro and in vivo assays. Effects of MC-LR (1, 10, 100, 500, 1000 and 5000 µg/L) on steroidogenesis were assessed in the H295R cells after 48 h. The contents of 17ß-estradiol (E2) and testosterone (T) increased in a non-dose-dependent manner, which showed positive correlations with the expression of steroidogenic genes. In the in vivo assay, adult male zebrafish were exposed to 0.3, 1, 3, 10 and 30 µg/L MC-LR for 30 d. Similarly, E2 and T contents in the testis were increased, accompanied by extensive up-regulation of steroidogenic genes, especially cyp19a. Meanwhile, the percentage of spermatid in the testis declined. In the liver, the vtg1 gene was significantly up-regulated while both the transcriptional and protein levels of the estrogenic receptor (ER) declined. These results indicate that MC-LR induced non-dose-dependent estrogenic effects at environmental concentrations, which may result from steroidogenesis stimulation via a non-ER-mediated pathway. Our findings support a paradigm shift in the risk assessment of MC-LR from traditional toxicity to estrogenic risk, particularly at low concentrations, and emphasize the potential threat to the male reproductive capacity of wildlife in bloom areas.


Subject(s)
Estrogens/toxicity , Gonadal Steroid Hormones/metabolism , Microcystins/toxicity , Reproduction/drug effects , Water Pollutants, Chemical/toxicity , Animals , Aromatase/biosynthesis , Cell Line , Estradiol/metabolism , Estrone/metabolism , Humans , Liver/metabolism , Male , Marine Toxins , Receptors, Estrogen/metabolism , Testis/drug effects , Testosterone/metabolism , Up-Regulation/drug effects , Zebrafish/metabolism , Zebrafish Proteins/biosynthesis
14.
Comp Biochem Physiol B Biochem Mol Biol ; 221-222: 11-17, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29655871

ABSTRACT

Brain aromatase is a key enzyme exclusively expressed in fish radial glial cells that convert androgens into estrogens, thus controlling neuroendocrine functions and neurogenesis. As an important step in characterizing the neuroendocrine systems of Rhamdia quelen (jundiá), a partial cDNA sequence (1045 bp) of brain aromatase (cyp19a1b) was cloned and sequenced. At the nucleotide level the cDNA sequence was found to be 88% identical to cyp19a1b of two species of catfish, Ictalurus punctatus and Silurus meridionalis. The predicted amino acid sequence was between 80 and 91% similar to other teleosts. Real-time RT-qPCR analysis revealed that cyp19a1b was detected in pituitary, hypothalamus, telencephalon, head and posterior kidneys, liver and gonads (testis and ovary) of both males and females. The effects of E2 on cyp19a1b expression are sexually dimorphic in R. quelen. The injection of 17ß-estradiol (E2) decreased head kidney mRNA levels of cyp19a1b in males and increased cyp19a1b mRNA levels in the pituitary and head kidney of females. This study demonstrated that the R. quelen cyp19a1b gene is expressed in brain, pituitary and peripheral tissues in both males and females.


Subject(s)
Aromatase , Catfishes , Cloning, Molecular , Fish Proteins , Gene Expression Regulation, Developmental/physiology , Sequence Analysis, DNA , Animals , Aromatase/biosynthesis , Aromatase/genetics , Catfishes/genetics , Catfishes/metabolism , Fish Proteins/biosynthesis , Fish Proteins/genetics , Organ Specificity
15.
FASEB J ; 32(6): 3361-3370, 2018 06.
Article in English | MEDLINE | ID: mdl-29401621

ABSTRACT

White adipose tissue inflammation is linked with increased aromatase gene expression and estrogen production, a major risk factor for breast cancer in obese postmenopausal women. TNF-α, a proinflammatory cytokine, is a key driver of aromatase promoter I.4-mediated expression in adipose tissue. In this study, we have shown that IL-10, an anti-inflammatory cytokine, suppressed both TNF-α-stimulated human aromatase reporter-luciferase (hARO-Luc) expression in mouse bone marrow mesenchymal stromal cells and aromatase gene expression in human breast adipose stromal cells (ASCs). IL-10 blocked TNF-α-stimulated ERK1/2 activation in ASCs, suggesting an inhibitory effect through the MAPK signaling pathway. The links among obesity, IL-10, and aromatase were confirmed in ovariectomized (OVX) hARO-Luc mice, where increased adiposity was associated with upregulation of aromatase reporter activity and reduced IL-10 level in the mammary fat pad. OVX mice also exhibited changes in gut microbiota, similar to that in obese women, indicating altered immune function. In summary, our results suggest that increased adiposity, induced by the lack of ovarian hormones, results in enhanced expression of aromatase in mammary adipose tissue, mediated by reduction in local IL-10. These findings may bring new insights into the mechanisms involved in the development of postmenopausal breast cancer, as well as novel approaches for prevention.-Martínez-Chacón, G., Brown, K. A., Docanto, M. M., Kumar, H., Salminen, S., Saarinen, N., Mäkelä, S. IL-10 suppresses TNF-α-induced expression of human aromatase gene in mammary adipose tissue.


Subject(s)
Adipose Tissue/enzymology , Aromatase/biosynthesis , Breast/enzymology , Gene Expression Regulation, Enzymologic , Interleukin-10/metabolism , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Animals , Female , Humans , Mammary Glands, Animal/enzymology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
16.
J Cell Physiol ; 233(3): 1959-1970, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28294325

ABSTRACT

Granulosa cell (GC) culture models mimicking the intrafollicular environment are limited. Such models have a great potential in reproductive toxicity studies. The buffalo, a monovulatory species like humans, could be a better model than polyovulatory rodents. Therefore, we targeted the development and characterization of three-dimensional (3D) culture systems for buffalo GCs. The GCs from small ovarian follicles (SF) maintained the CYP19 gene expression for 144 hr in a 2D culture system. Hence, GCs from SF were cultured directly in 3D using hanging drop and Poly-([2-hydroxyethyl methacrylate]) (polyHEMA) methods in the DMEM media containing 1 ng/ml FSH and 10 ng/ml IGF-1 for 144 hr. The expression profile of nine GC-specific transcripts; CYP19, TNFAIP6, AMH, PTI, NR4A1, FSHR, RUNX, LHR, and COX2/PTGS2; revealed that 3D-spheroids developed in hanging drop method maintained the GC phenotype of preovulatory follicles. Therefore, hanging drop method is a best method for culturing GCs to mimic the intrafollicular environment.


Subject(s)
Aromatase/biosynthesis , Cell Culture Techniques/methods , Granulosa Cells/metabolism , Spheroids, Cellular/metabolism , Animals , Aromatase/genetics , Buffaloes , Cells, Cultured , Female , Follicle Stimulating Hormone/pharmacology , Insulin-Like Growth Factor I/pharmacology , Methacrylates/pharmacology
17.
J Chem Neuroanat ; 90: 70-79, 2018 07.
Article in English | MEDLINE | ID: mdl-29288708

ABSTRACT

Aromatase is a steroidogenic enzyme involved in the conversion of testosterone into estradiol. Teleosts are unique among vertebrates in possessing two distinct aromatase genes that show different expression patterns within the body. Since the brain is the essential organ underlying the control of behavior, an understanding of the expression pattern of aromatase in the brain can help to identify neural circuits and behaviors that are most likely to be affected by aromatase activity. In addition, identifying species differences in aromatase expression in the brain can further our understanding of divergence in behaviors regulated by local estradiol production and estrogen signaling. Apteronotus leptorhynchus is a species of weakly electric fish in which little is known about sex steroid expression within the brain and its role in electric signaling behavior. The goal of this study was to identify the mRNA expression pattern of aromatase in the brain of A. leptorhynchus. Aromatase mRNA was detected in several parts of the forebrain and in the pituitary gland; however, no aromatase expression was detected in the midbrain or hindbrain. These findings in A. leptorhynchus support a role for aromatase activity in reproduction, but no direct role in electric signaling behavior in non-breeding fish. The findings of this study help to broaden the basis for making phylogenetic comparisons of aromatase expression across teleost lineages as well as different signaling systems, and provide information on behaviors and neural circuits that are potentially affected by local estradiol production in A. leptorhynchus.


Subject(s)
Aromatase/biosynthesis , Brain/enzymology , Electric Fish/metabolism , Animals , Aromatase/analysis , RNA, Messenger/analysis
18.
Steroids ; 128: 114-119, 2017 12.
Article in English | MEDLINE | ID: mdl-28951168

ABSTRACT

Placenta produces progesterone and estradiol for maintaining pregnancy. Two critical enzymes are responsible for their production: 3ß-hydroxysteroid dehydrogenase 1 (HSD3B1) that catalyzes the formation of progesterone from pregnenolone and aromatase that catalyzes the production of estradiol from testosterone. Fungicide ziram may disrupt the placental steroid production. In the present study, we investigated the effects of ziram on steroid formation in human placental cell line JEG-3 cells and on HSD3B1 and aromatase in the human placenta. Ziram did not inhibit progesterone production in JEG-3 cells and HSD3B1 activity at 100µM. Ziram was a potent aromatase inhibitor with the half maximal inhibitory concentration (IC50) value of 333.8nM. When testosterone was used to determine the mode of action, ziram was found to be a competitive inhibitor. Docking study showed that ziram binds to the testosterone binding pocket of the aromatase. In conclusion, ziram is a potent inhibitor of human aromatase.


Subject(s)
Aromatase Inhibitors/chemistry , Aromatase/genetics , Multienzyme Complexes/genetics , Placenta/metabolism , Progesterone Reductase/genetics , Steroid Isomerases/genetics , Ziram/chemistry , Aromatase/biosynthesis , Aromatase/chemistry , Aromatase Inhibitors/therapeutic use , Cell Line, Tumor , Estradiol/metabolism , Female , Humans , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/chemistry , Placenta/chemistry , Placenta/drug effects , Pregnancy , Pregnenolone/metabolism , Progesterone/biosynthesis , Progesterone Reductase/biosynthesis , Progesterone Reductase/chemistry , Protein Binding , Steroid Isomerases/biosynthesis , Steroid Isomerases/chemistry , Testosterone/metabolism , Ziram/therapeutic use
19.
Sci Rep ; 7(1): 5320, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28706210

ABSTRACT

During development sex differences in aromatase expression in limbic regions of mouse brain depend on sex chromosome factors. Genes on the sex chromosomes may affect the hormonal regulation of aromatase expression and this study was undertaken to explore that possibility. Male E15 anterior amygdala neuronal cultures expressed higher levels of aromatase (mRNA and protein) than female cultures. Furthermore, treatment with oestradiol (E2) or dihydrotestosterone (DHT) increased Cyp19a1 expression and aromatase protein levels only in female neuronal cultures. The effect of E2 on aromatase expression was not imitated by oestrogen receptor (ER) α agonist PPT or the GPER agonist G1, but it was fully reproduced by DPN, a specific ligand of ERß. By contrast, the effect of DHT on aromatase expression was not blocked by the anti-androgen flutamide, but completely abrogated by the ERß antagonist PHTPP. Experiments using the four core genotype model showed a sex chromosome effect in ERß expression (XY > XX) and regulation by E2 or DHT (only XX respond) in amygdala neurons. In conclusion, sex chromosome complement governs the hormonal regulation of aromatase expression through activation of ERß in developing mouse brain.


Subject(s)
Amygdala/embryology , Amygdala/enzymology , Aromatase/biosynthesis , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Developmental , Neurons/enzymology , Sex Chromosomes , Animals , Cells, Cultured , Dihydrotestosterone/metabolism , Estradiol/metabolism , Female , Male , Mice
20.
Dev Biol ; 428(1): 232-244, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28606561

ABSTRACT

Macrophages are required for proper mammary gland development and maintaining tissue homeostasis. However, the mechanisms by which macrophages regulate this process remain unclear. Here, we identify STAT5 as an important regulator of macrophage function in the developing mammary gland. Analysis of mammary glands from mice with STAT5-deficient macrophages demonstrates delayed ductal elongation, enhanced ductal branching and increased epithelial proliferation. Further analysis reveals that STAT5 deletion in macrophages leads to enhanced expression of proliferative factors such as Cyp19a1/aromatase and IL-6. Mechanistic studies demonstrate that STAT5 binds directly to the Cyp19a1 promoter in macrophages to suppress gene expression and that loss of STAT5 results in enhanced stromal expression of aromatase. Finally, we demonstrate that STAT5 deletion in macrophages cooperates with oncogenic initiation in mammary epithelium to accelerate the formation of estrogen receptor (ER)-positive hyperplasias. These studies establish the importance of STAT5 in macrophages during ductal morphogenesis in the mammary gland and demonstrate that altering STAT5 function in macrophages can affect the development of tissue-specific disease.


Subject(s)
Macrophage Activation/genetics , Macrophages/cytology , Mammary Glands, Animal/embryology , Morphogenesis/genetics , Organogenesis/genetics , STAT5 Transcription Factor/genetics , Animals , Aromatase/biosynthesis , Aromatase/genetics , Cell Line , Cell Proliferation/genetics , Enzyme Activation/genetics , Epithelial Cells/cytology , Female , Gene Expression Regulation/genetics , Interleukin-6/biosynthesis , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...