Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 83
1.
Clin Cancer Res ; 29(24): 5217-5226, 2023 12 15.
Article En | MEDLINE | ID: mdl-37888299

PURPOSE: In estrogen receptor-positive (ER+) breast cancer, single-nucleotide polymorphisms (SNP) in the aromatase gene might affect aromatase inhibitors (AI) metabolism and efficacy. Here, we assessed the impact of SNP on prognosis and toxicity of patients receiving adjuvant letrozole. EXPERIMENTAL DESIGN: We enrolled 886 postmenopausal patients in the study. They were treated with letrozole for 2 to 5 years after taking tamoxifen for 2 to 6 years, continuing until they completed 5 to 10 years of therapy. Germline DNA was genotyped for SNP rs4646, rs10046, rs749292, and rs727479. Log-rank test and Cox model were used for disease-free survival (DFS) and overall survival (OS). Cumulative incidence (CI) of breast cancer metastasis was assessed through competing risk analysis, with contralateral breast cancer, second malignancies and non-breast cancer death as competing events. CI of skeletal and cardiovascular events were assessed using DFS events as competing events. Subdistribution HR (sHR) with 95% confidence intervals were calculated through Fine-Gray method. RESULTS: No SNP was associated with DFS. Variants rs10046 [sHR 2.03, (1.04-2.94)], rs749292 [sHR 2.11, (1.12-3.94)], and rs727479 [sHR 2.62, (1.17-5.83)] were associated with breast cancer metastasis. Three groups were identified on the basis of the number of these variants (0, 1, >1). Variant-based groups were associated with breast cancer metastasis (10-year CI 2.5%, 7.6%, 10.7%, P = 0.035) and OS (10-year estimates 96.5%, 93.0%, 89.6%, P = 0.030). Co-occurrence of rs10046 and rs749292 was negatively associated with 10-year CI of skeletal events (3.2% vs. 10%, P = 0.033). A similar association emerged between rs727479 and cardiovascular events (0.3% vs. 2.1%, P = 0.026). CONCLUSIONS: SNP of aromatase gene predict risk of metastasis and AI-related toxicity in ER+ early breast cancer, opening an opportunity for better treatment individualization.


Aromatase Inhibitors , Breast Neoplasms , Female , Humans , Aromatase/genetics , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/toxicity , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/genetics , Chemotherapy, Adjuvant , Letrozole/adverse effects , Polymorphism, Single Nucleotide , Tamoxifen/therapeutic use
2.
Toxicol In Vitro ; 81: 105332, 2022 Jun.
Article En | MEDLINE | ID: mdl-35176449

Human aromatase, also called CYP19A1, plays a major role in the conversion of androgens into estrogens. Inhibition of aromatase is an important target for estrogen receptor (ER)-responsive breast cancer therapy. Use of azole compounds as aromatase inhibitors is widespread despite their low selectivity. A toxicological evaluation of commonly used azole-based drugs and agrochemicals with respect to CYP19A1 is currently requested by the European Union- Registration, Evaluation, Authorization and Restriction of Chemicals (EU-REACH) regulations due to their potential as endocrine disruptors. In this connection, identification of structural alerts (SAs) is an effective strategy for the toxicological assessment and safe drug design. The present study describes the identification of SAs of azole-based chemicals as guiding experts to predict the aromatase activity. Total 21 SAs associated with aromatase activity were extracted from dataset of 326 azole-based drugs/chemicals obtained from Tox21 library. A cross-validated classification model having high accuracy (error rate 5%) was proposed which can precisely classify azole chemicals into active/inactive toward aromatase. In addition, mechanistic details and toxicological properties (agonism/antagonism) of azoles with respect to aromatase were explored by comparing active and inactive chemicals using structure-activity relationships (SAR). Lastly, few structural alerts were applied to form chemical categories for read-across applications.


Aromatase , Azoles , Aromatase/metabolism , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/toxicity , Azoles/toxicity , Cytochrome P-450 CYP1A1 , Humans , Receptors, Estrogen , Structure-Activity Relationship
3.
Reprod Toxicol ; 108: 43-55, 2022 03.
Article En | MEDLINE | ID: mdl-35091028

The development and application of (quantitative) structure-activity relationship ((Q)SAR) models for reproductive toxicology remains challenging, given the complexity of the endpoint and the risks associated with subsequent decision making. Adverse outcome pathways (AOPs) organise knowledge and provide context of model outputs, aiding risk assessors' decision making. Using aromatase as an example, we demonstrate how AOPs can be used to contextualise a variety of (Q)SAR approaches. AOPs stemming from aromatase inhibition - leading to adverse outcomes of regulatory significance - were synthesised and annotated with relevant assays, assay data and (Q)SAR models. The resulting framework enabled the deployment of different types of (Q)SAR models that predict for key events along the pathway. The use of models for molecular initiating events enables relevant knowledge to span a wider area of chemical space - compared to using models trained solely on in vivo toxicity data. Utilising such methods, alongside additional assay data and exposure information, could lead to improved risk assessment strategies during compound prioritisation and labelling.


Adverse Outcome Pathways , Aromatase Inhibitors/toxicity , Quantitative Structure-Activity Relationship , Reproduction/drug effects , Animals , Aromatase Inhibitors/chemistry , Humans
4.
Eur J Histochem ; 65(3)2021 Sep 03.
Article En | MEDLINE | ID: mdl-34474552

The epididymis is an organ that plays a key role in sperm maturation. The aim of this study was to examine the association between the chronic treatment of mature male rats with letrozole and morphological evaluation and morphometric values of epididymis as well as changes in the number of apoptotic cells in epididymal epithelium. Adult rats were treated with letrozole for 6 months and the epididymis weight, morphology, morphometric values and the number of apoptotic cells in  the epithelium were examined. Long-term aromatase inhibition resulted in presence of intraepithelial clear vacuoles, hyperplasia of clear cells and a hyperplastic alteration in the epithelium known as a cribriform change. Moreover, changes in diameters of the epididymal duct and the epididymal lumen and changes in the epididymal epithelium height were observed. The number of apoptotic epithelial cells was increased in letrozole-treated group. It can be indicated that chronic treatment with letrozole can affect morphology, morphometric values and apoptosis in the epididymis of adult male rats. Observed changes are similar to that observed in the aging processes and may also be important for patients treated with aromatase inhibitors.


Apoptosis/drug effects , Aromatase Inhibitors/toxicity , Epididymis/drug effects , Epithelium/drug effects , Letrozole/toxicity , Animals , Epididymis/metabolism , Epithelium/metabolism , Estradiol/metabolism , Male , Rats, Wistar
5.
Toxicol Appl Pharmacol ; 425: 115600, 2021 08 15.
Article En | MEDLINE | ID: mdl-34081940

Among postmenopausal women with estrogen receptor-positive breast cancer, more than 80% receive hormone therapy including aromatase inhibitors (AIs). Half of them develop chronic arthralgia - characterized by symmetric articular pain, carpal tunnel syndrome, morning stiffness, myalgia and a decrease in grip strength - which is associated with treatment discontinuation. Only a few animal studies have linked AI treatment to nociception, and none to arthralgia. Thus, we developed a new chronic AI-induced nociceptive disorder model mimicking clinical symptoms induced by AIs, using subcutaneous letrozole pellets in ovariectomized (OVX) rats. Following plasma letrozole dosage at the end of the experiment (day 73), only rats with at least 90 ng/ml of letrozole were considered significantly exposed to letrozole (OVX + high LTZ group), whereas treated animals with less than 90 ng/ml were pooled in the OVX + low LTZ group. Chronic nociceptive disorder set in rapidly and was maintained for more than 70 days in the OVX + high LTZ group. Furthermore, OVX + high LTZ rats saw no alteration in locomotion, myalgia or experimental anxiety during this period. Bone parameters of the femora were significantly altered in all OVX rats compared to Sham+vehicle pellet. A mechanistic analysis focused on TRPA1, receptor suspected to mediate AI-evoked pain, and showed no modification in its expression in the DRG. This new long-lasting chronic rat model, efficiently reproduces the symptoms of AI-induced nociceptive disorder affecting patients' daily activities and quality-of-life. It should help to study the pathophysiology of this disorder and to promote the development of new therapeutic strategies.


Aromatase Inhibitors/toxicity , Disease Models, Animal , Letrozole/toxicity , Nociception/drug effects , Animals , Body Weight/drug effects , Chronic Disease , Female , Ganglia, Spinal , Gene Expression Regulation/drug effects , Motor Activity/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley
6.
J Ethnopharmacol ; 278: 114318, 2021 Oct 05.
Article En | MEDLINE | ID: mdl-34111539

ETHNOPHARMACOLOGICAL RELEVANCE: Chickpea was used in both greek and indian traditional medicine for hormonal related conditions as menstrual induction, acceleration of parturation, treatment of retained placenta and stimulation of lactation. Chickpea (Cicer arietinum) sprout isoflavone isolates exhibited reasonable estrogenic activities. Isoflavones, a subtype of phytoestrogens, are plant derivatives with moderate estrogenic activity that tend to have protective effects on hormonal and metabolic abnormalities of women with polycystic ovary syndrome (PCOS). AIM OF THE STUDY: In this study, we investigated the effect of UPLC/ESI-MS characterized Cicer arietinum L. seeds ethanol extract (CSE) on ovarian hormones, oxidative response and ovarian histological changes on induced PCOS rat model. MATERIALS AND METHODS: Thirty-five rats were divided into five groups including negative control, PCOS, and treatment groups. PCOS was induced using letrozole (1 mg/kg) daily orally for 21 days. Each treatment group was treated with one of the following for 28 days after induction of PCOS: clomiphene citrate (1 mg/kg), and CSE at 250 and 500 mg/kg. Ovaries and uteri were excised, weighed and their sections were used for quantitative real-time reverse transcriptase polymerase chain reaction, antioxidant assays and histomorphometric study of the ovaries. The antioxidant assays, histopathological examination, hormonal and metabolic profiles, and Cyp11a1(steroidogenic enzyme) mRNA expression were measured. RESULTS: In all treatment groups, ovarian weight was significantly decreased despite having no significant effect on uterine weight. Histomorphometric study in the treatment groups revealed a significant decrease in the number and diameter of cystic follicles, a significant increase in granulosa cell thickness while, thickness of theca cells was significantly decreased when compared to PCOS. Hormone levels, metabolic profile and antioxidant status were improved in the treatment groups. Moreover, Cyp11a1 mRNA expression was significantly downregulated in the treatment groups compared to PCOS. CONCLUSIONS: In the current study, CSE enhanced the reproductive and metabolic disorders which were associated with PCOS induction. For the first time, we have highlighted the effect of CSE in treating PCOS and its associated manifestations.


Cicer/chemistry , Letrozole/toxicity , Phytotherapy , Plant Extracts/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Animals , Aromatase Inhibitors/toxicity , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Clomiphene/therapeutic use , Dose-Response Relationship, Drug , Estrogen Antagonists/therapeutic use , Female , Gene Expression Regulation, Enzymologic/drug effects , Organ Size , Ovary/pathology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Polycystic Ovary Syndrome/chemically induced , Random Allocation , Rats
7.
Toxicol Mech Methods ; 31(7): 538-545, 2021 Sep.
Article En | MEDLINE | ID: mdl-34036875

Radiation-induced pulmonary fibrosis (RIPF) is a known complication in cancer patients after getting thoracic radiotherapy. Aromatase inhibitors (AIs) as anastrozole have been used instead of tamoxifen for adjuvant endocrine treatment of postmenopausal women with hormone sensitive breast cancer. This study is to evaluate the concurrent treatment of anastrozole and RIPF in rats. Twenty four female Wistar rats were distributed into 4 groups: Control (C), Radiation group (R) (total dose 30 Gy in 10 fractions, 5 fractions/week), anastrozole group (A) (0.003 mg/200 g body weight) orally for 14 consecutive days, and Radiation + anastrozole group (R + A). Radiation exposure resulted in a significant increase (p < 0.05) in pulmonary Transforming growth factor-beta 1 (TGF-ß), SMAD family member 3 (Smad3), Platelet-derived growth factor (PDGF), malondialdehyde (MDA), Total nitrate/nitrite (NO), interleukin 1ß (IL-1ß) and interleukin 6 (IL-6) compared to the control group. While, significant decreases (p < 0.05) in superoxide dismutase (SOD) activity, reduced glutathione (GSH) and connective tissue growth factor (CTGF) were observed in lung tissue. These alterations were minimized by anastrozole intervention. Also, anastrozole markedly hindered the lung histopathological changes observed after radiation. Concomitant use of anastrozole with radiation seems to attenuate radiation-induced pulmonary toxicity via TGF-ß/Smad 3 and TGF-ß/PDGF pathways in rats.


Pulmonary Fibrosis , Signal Transduction , Animals , Aromatase Inhibitors/toxicity , Female , Lung , Platelet-Derived Growth Factor , Pulmonary Fibrosis/chemically induced , Rats , Rats, Wistar , Smad3 Protein , Transforming Growth Factor beta
8.
Gynecol Endocrinol ; 37(4): 337-341, 2021 Apr.
Article En | MEDLINE | ID: mdl-32851887

AIMS: The effective treatment of polycystic ovary syndrome (PCOS)-related hormonal disorders necessitates the development of novel treatment strategies. Resveratrol is found in certain food products, and is known to exhibit phytoestrogen properties. The present study was to assess whether resveratrol exhibits beneficial phytoestrogenic effects and associated hormonal modulation in a rat model of PCOS. MATERIALS AND METHODS: This model was established by administering oral letrozole to female Sprague-Dawley (SD) rats prior to randomizing them into control, model and resveratrol treatment groups (40, 80, or 160 mg/kg). Animals were treated for 30 days, after which time ovarian tissues were collected and evaluated via hematoxylin and eosin staining. In addition, serum levels of estradiol and adiponectin were assessed via ELISA, and ovarian expression of nesfatin-1 and aromatase was assessed through RT-PCR and western blotting. RESULTS: We found that resveratrol administration was associated with increased levels of plasma adiponectin and estradiol levels and restoration of normal ovarian morphology in PCOS model animals. In addition, this treatment was linked to the increased ovarian expression of nesfatin-1 and aromatase at the RNA and protein levels. CONCLUSIONS: Together things findings suggest that resveratrol may represent an effective tool for treating PCOS owing to its phytoestrogenic properties.


Ovary/drug effects , Phytoestrogens/pharmacology , Polycystic Ovary Syndrome/pathology , Resveratrol/pharmacology , Adiponectin/metabolism , Animals , Aromatase/drug effects , Aromatase/genetics , Aromatase Inhibitors/toxicity , Disease Models, Animal , Estradiol/metabolism , Female , Letrozole/toxicity , Nucleobindins/drug effects , Nucleobindins/genetics , Ovary/metabolism , Ovary/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Random Allocation , Rats
9.
Environ Toxicol Chem ; 39(4): 913-922, 2020 04.
Article En | MEDLINE | ID: mdl-31965587

Predictive approaches to assessing the toxicity of contaminant mixtures have been largely limited to chemicals that exert effects through the same biological molecular initiating event. However, by understanding specific pathways through which chemicals exert effects, it may be possible to identify shared "downstream" nodes as the basis for forecasting interactive effects of chemicals with different molecular initiating events. Adverse outcome pathway (AOP) networks conceptually support this type of analysis. We assessed the utility of a simple AOP network for predicting the effects of mixtures of an aromatase inhibitor (fadrozole) and an androgen receptor agonist (17ß-trenbolone) on aspects of reproductive endocrine function in female fathead minnows. The fish were exposed to multiple concentrations of fadrozole and 17ß-trenbolone individually or in combination for 48 or 96 h. Effects on 2 shared nodes in the AOP network, plasma 17ß-estradiol (E2) concentration and vitellogenin (VTG) production (measured as hepatic vtg transcripts) responded as anticipated to fadrozole alone but were minimally impacted by 17ß-trenbolone alone. Overall, there were indications that 17ß-trenbolone enhanced decreases in E2 and vtg in fadrozole-exposed fish, as anticipated, but the results often were not statistically significant. Failure to consistently observe hypothesized interactions between fadrozole and 17ß-trenbolone could be due to several factors, including lack of impact of 17ß-trenbolone, inherent biological variability in the endpoints assessed, and/or an incomplete understanding of interactions (including feedback) between different pathways within the hypothalamic-pituitary-gonadal axis. Environ Toxicol Chem 2020;39:913-922. © 2020 SETAC.


Adverse Outcome Pathways , Androgens/toxicity , Aromatase Inhibitors/toxicity , Cyprinidae/physiology , Endocrine System/drug effects , Reproduction/drug effects , Animals , Cyprinidae/metabolism , Drug Synergism , Estradiol/metabolism , Fadrozole/toxicity , Female , Hypothalamo-Hypophyseal System/drug effects , Male , Ovary/drug effects , Ovary/metabolism , Trenbolone Acetate/toxicity , Vitellogenins/metabolism
10.
Regul Toxicol Pharmacol ; 109: 104510, 2019 Dec.
Article En | MEDLINE | ID: mdl-31676319

Synthesis of 11 steroid hormones in human adrenocortical carcinoma cells (H295R) was measured in a high-throughput steroidogenesis assay (HT-H295R) for 656 chemicals in concentration-response as part of the US Environmental Protection Agency's ToxCast program. This work extends previous analysis of the HT-H295R dataset and model by examining the utility of a novel prioritization metric based on the Mahalanobis distance that reduced these 11-dimensional data to 1-dimension via calculation of a mean Mahalanobis distance (mMd) at each chemical concentration screened for all hormone measures available. Herein, we evaluated the robustness of mMd values, and demonstrate that covariance and variance of the hormones measured appear independent of the chemicals screened and are inherent to the assay; the Type I error rate of the mMd method is less than 1%; and, absolute fold changes (up or down) of 1.5 to 2-fold have sufficient power for statistical significance. As a case study, we examined hormone responses for aromatase inhibitors in the HT-H295R assay and found high concordance with other ToxCast assays for known aromatase inhibitors. Finally, we used mMd and other ToxCast cytotoxicity data to demonstrate prioritization of the most selective and active chemicals as candidates for further in vitro or in silico screening.


Aromatase Inhibitors/toxicity , Endocrine Disruptors/toxicity , High-Throughput Screening Assays/methods , Steroids/biosynthesis , Cell Line, Tumor , Data Interpretation, Statistical , Datasets as Topic , False Positive Reactions , High-Throughput Screening Assays/standards , Humans , Reproducibility of Results , Toxicity Tests/methods , Toxicity Tests/standards , United States , United States Environmental Protection Agency/standards
11.
Toxicology ; 425: 152247, 2019 09 01.
Article En | MEDLINE | ID: mdl-31330226

Azole antifungal drugs are used worldwide to treat a variety of fungal infections such as vulvovaginal candidiasis, particularly in pregnant women who are at increased risk. The aim of this study was to mechanistically investigate the endocrine disrupting potential of four commonly used azole antifungal drugs; clotrimazole, miconazole, ketoconazole and fluconazole in vitro using the H295R cell assay and two recombinant, CYP17A1 and CYP19A1 (aromatase), assays. Steroids were quantified using LC-MS/MS. In both recombinant assays, all four azoles inhibited the CYP enzymes investigated, at therapeutically relevant concentrations. However, responses were much more complex in the H295R cell line. Clotrimazole inhibited steroid production in a dose-dependent manner with IC50 values for CYP17A1 and CYP19A1 in the range 0.017-0.184 µM. Miconazole and ketoconazole increased all steroids on the hydroxylase axis (IC50 MIC: 0.042-0.082 µM, KET: 0.041-1.2 µM), leading to accumulation of progestagens and corticosteroids and suppression of androgens and estrogens, indicating inhibition of CYP17A1, in particular lyase activity. However, ketoconazole suppressed all steroids at higher concentrations, resulting in bell-shaped curves for all steroids on the hydroxylase axis. Fluconazole was found to inhibit CYP17A1-lyase activity, causing suppression of androgens (IC50 = 114-209 µM) and estrogens (IC50 = 28 µM). The results indicate that these four azole drugs are highly potent in vitro and, based on plasma Cmax values, may exert endocrine disrupting effects at therapeutically relevant concentrations. This raises concern for endocrine related effects in patients using azole antifungal drugs, particularly when taken during sensitive periods like pregnancy.


Antifungal Agents/toxicity , Aromatase/drug effects , Clotrimazole/toxicity , Endocrine Disruptors/toxicity , Fluconazole/toxicity , Ketoconazole/toxicity , Miconazole/toxicity , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Aromatase Inhibitors/toxicity , Cell Line, Tumor , Dose-Response Relationship, Drug , Gas Chromatography-Mass Spectrometry , Humans , Inhibitory Concentration 50
12.
Sci Rep ; 9(1): 6599, 2019 04 29.
Article En | MEDLINE | ID: mdl-31036921

The fish short-term reproduction assay (FSTRA) is a common in vivo screening assay for assessing endocrine effects of chemicals on reproduction in fish. However, the current reliance on measures such as egg number, plasma vitellogenin concentration and morphological changes to determine endocrine effects can lead to false labelling of chemicals with non-endocrine modes- of-action. Here, we integrated quantitative liver and gonad shotgun proteomics into the FSTRA in order to investigate the causal link between an endocrine mode-of-action and adverse effects assigned to the endocrine axis. Therefore, we analyzed the molecular effects of fadrozole-induced aromatase inhibition in zebrafish (Danio rerio). We observed a concentration-dependent decrease in fecundity, a reduction in plasma vitellogenin concentrations and a mild oocyte atresia with oocyte membrane folding in females. Consistent with these apical measures, proteomics revealed a significant dysregulation of proteins involved in steroid hormone secretion and estrogen stimulus in the female liver. In the ovary, the deregulation of estrogen synthesis and binding of sperm to zona pellucida were among the most significantly perturbed pathways. A significant deregulation of proteins targeting the transcriptional activity of estrogen receptor (esr1) was observed in male liver and testis. Our results support that organ- and sex-specific quantitative proteomics represent a promising tool for identifying early gene expression changes preceding chemical-induced adverse outcomes. These data can help to establish consistency in chemical classification and labelling.


Endocrine System/drug effects , Estrogen Receptor alpha/genetics , Proteomics , Water Pollutants, Chemical/toxicity , Zebrafish Proteins/genetics , Animals , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/toxicity , Estrogens/metabolism , Fadrozole/pharmacology , Fadrozole/toxicity , Female , Gene Expression Regulation, Developmental/drug effects , Gonadal Steroid Hormones/antagonists & inhibitors , Gonadal Steroid Hormones/biosynthesis , Gonads/drug effects , Gonads/metabolism , Liver/drug effects , Liver/metabolism , Male , Reproduction/drug effects , Testis/drug effects , Water Pollutants, Chemical/pharmacology , Zebrafish/genetics , Zebrafish/growth & development
13.
Environ Toxicol Pharmacol ; 68: 133-140, 2019 May.
Article En | MEDLINE | ID: mdl-30889543

Acute developmental exposure to pharmaceuticals or environmental contaminants can have deleterious, long lasting effects. Many of these compounds are endocrine disruptors (EDCs) that target estrogen signaling, with effects on reproductive and non-reproductive tissues. We recently reported that zebrafish larvae transiently exposed to the pharmaceutical EDC 4-OH-A display visual deficits as adults. Here, we examine whether these long-term effects are due to compound-induced morphological and/or cellular changes. Zebrafish aged 24 h, 48 h, 72 h, or 7 days post-fertilization (larvae) or 3-4mos (adults) were exposed to either 4-OH-A or PCB1254 for 24 h. After that time, notochord length, eye diameter, inter-eye distance, and heart rate were measured from larvae; and aromatase (estrogen synthase) activity was measured in homogenates of adult brain tissue. In general, indices of larval growth and development were not altered by 24 h exposure to either compound. 4-OH-A potently inhibited aromatase activity, while PCB1254 did not, with inhibition continuing even after removal from treatment. These results support differential function of EDCs and indicate that developmental exposure to 4-OH-A causes sustained inhibition of aromatase, which could be associated with altered adult behaviors.


Androstenedione/analogs & derivatives , Aromatase Inhibitors/toxicity , Endocrine Disruptors/toxicity , Zebrafish , Androstenedione/toxicity , Animals , Aromatase/metabolism , Brain/drug effects , Brain/metabolism , Larva/growth & development , Larva/metabolism , Zebrafish/growth & development , Zebrafish/metabolism
14.
Food Chem Toxicol ; 128: 46-53, 2019 Jun.
Article En | MEDLINE | ID: mdl-30922969

Human placental CYP19A1 catalyzes the estrogen synthesis from androgens. The enzyme is encoded by CYP19A1 gene located in chromosome 15q21. This enzyme is a monooxygenase in the smooth endoplasmic reticulum. The various promoters of the CYP19A1 gene determine its expression in different tissues and the distal promoter I.1 controls its expression in the placenta and retinoids can regulate the expression. Many food components and environmental chemicals inhibit CYP19A1 activity via different modes of action. These chemicals include gossypol, flavones, flavanones, chalconoids, resveratrol, and tobacco alkaloids derived from foods as well as phthalates, insecticides, fungicides, and biocides in the contaminated foods. The inhibition of placental CYP19A1 could impair pregnancy.


Aromatase Inhibitors/toxicity , Aromatase/drug effects , Environmental Pollutants/toxicity , Estrogen Receptor Modulators/toxicity , Food Analysis , Placenta/drug effects , Aromatase/chemistry , Aromatase/genetics , Estrogen Receptor Modulators/chemistry , Female , Humans , Placenta/enzymology , Pregnancy , Promoter Regions, Genetic/drug effects
15.
Environ Pollut ; 244: 513-521, 2019 Jan.
Article En | MEDLINE | ID: mdl-30366299

BADGE (bisphenol A diglycidyl ether) is a synthesis product of bisphenol A (BPA), which, like other plasticizers, can cross the human placenta and reach the foetus. However, compared to BPA, there is almost no toxicological information. This work investigates the toxicity, endocrine and lipid disruption potential of BADGE and its hydrolysed and chlorinated derivatives (BADGE·H2O and BADGE·2HCl) in human placental JEG-3 cells. The analysis of culture medium by HPLC-ESI(+)-QqQ evidenced a good bioavailability of BADGE·2HCl and BADGE·H2O, but low stability of BADGE. Regardless, BADGE·2HCl and BADGE showed higher cytotoxicity than BADGE·H2O, which was the only compound that significantly inhibited CYP19 activity (IC50 49 ±â€¯5 µM). JEG-3 cells lipidome analyzed by FIA-ESI(+/-)-Orbitrap was significantly altered by exposure to BADGE·2HCl and BADGE at concentrations at the low µM range. BADGE·2HCl lead to a strong decrease of diacyl- and triacyl-glycerides (DGs,TGs) together with some membrane lipids, while BADGE lead to an accumulation of TGs. The results evidence the ability of BADGE and derivatives to affect placental lipid handling and to modulate placental CYP19 activity (BADGE·H2O) and highlights the need to monitor human exposure to these compounds, at least as intensely as BPA is monitored.


Aromatase Inhibitors/toxicity , Aromatase/metabolism , Benzhydryl Compounds/toxicity , Carcinogens/toxicity , Epoxy Compounds/toxicity , Phenols/toxicity , Placenta/cytology , Plasticizers/toxicity , Cell Line, Tumor , Chromatography, High Pressure Liquid/methods , Diglycerides/metabolism , Female , Humans , Membrane Lipids/metabolism , Placenta/drug effects , Pregnancy , Reactive Oxygen Species/metabolism , Triglycerides/metabolism
16.
Reproduction ; 155(2): 141-151, 2018 02.
Article En | MEDLINE | ID: mdl-29196492

Polycystic ovary syndrome (PCOS) is associated with hyperandrogenism, oligo-anovulation, polycystic ovaries and metabolic syndrome. Many researchers reported that PCOS often starts with menarche in adolescents. Presently available animal model focuses on ovarian but not metabolic features of PCOS. Therefore, we hypothesized that high-fat diet feeding to pre-pubertal female rats results in both reproductive and metabolic features of PCOS. Pre-pubertal female rats were divided into two groups: group I received normal pellet diet and group II received high-fat diet (HFD). In the letrozole study, adult female rats were divided into two groups: group I received 1% carboxy methyl cellulose and group II received 1 mg/kg letrozole orally. Oral glucose tolerance test, lipid profile, fasting glucose, insulin, estrus cycle, hormonal profile, ovary weight, luteinizing hormone (LH) receptor and follicle-stimulating hormone receptor expression were measured. Polycystic ovarian morphology was assessed through histopathological changes of ovary. Feeding of HFD gradually increase glucose intolerance and fasting insulin levels. Triglyceride level was higher in HFD study while total cholesterol level was higher in the letrozole study. Alteration in testosterone and estrogen levels was observed in both studies. LH receptor expression was upregulated only in HFD study. Histopathological changes like increase cystic follicle, diminished granulosa cell layer and thickened theca cell layer were observed in letrozole as well as HFD study. High-fat diet initiated at pre-puberty age in rats produces both metabolic disturbances and ovarian changes similar to that observed clinically in PCOS patients. Letrozole on the other hand induces change in ovarian structure and function.


Biomarkers/metabolism , Diet, High-Fat/adverse effects , Polycystic Ovary Syndrome/etiology , Polycystic Ovary Syndrome/pathology , Sexual Maturation/drug effects , Animals , Aromatase Inhibitors/toxicity , Female , Gene Expression Profiling , Letrozole , Nitriles/toxicity , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Sprague-Dawley , Triazoles/toxicity
17.
Cell Physiol Biochem ; 44(1): 66-84, 2017.
Article En | MEDLINE | ID: mdl-29131020

BACKGROUND/AIMS: Reports regarding the role of androgen in breast cancer (BC) are conflicting. Some studies suggest that androgen could lead to undesirable responses in the presence of certain BC tumor characteristics. We have shown that androgen induces C-X-C motif chemokine 12 (CXCL12) in BC cell lines. Our aim was to identify the mechanisms regulating the phenotypic effects of androgen-induced CXCL12 on Androgen Receptor (AR) positive BC cell lines. METHODS: We analyzed the expression of CXCL12 and its receptors with qPCR and ELISA and the role of Nuclear Receptor Coactivator 1 (NCOA1) in this effect. AR effects on the CXCL12 promoter was studied via Chromatin-immunoprecipitation. We also analyzed publically available data from The Cancer Genome Atlas to verify AR-CXCL12 interactions and to identify the effect or Aromatase Inhibitors (AI) therapy on CXCL12 expression and disease progression in AR positive cases. RESULTS: CXCL12 induction occurs only in AR-positive BC cell lines, possibly via an Androgen Response Element, upstream of the CXCL12 promoter. The steroid receptor co-regulator NCOA1 is critical for this effect. Androgen only induced the motility of p53-mutant BC cells T47D cells via upregulation of CXCR4 expression while they had no effect on wild-type p53 MCF-7 cells. Loss of CXCR4 expression and depletion of CXCL12 abolished the effect of androgen in T47D cells while inhibition of p53 expression in MCF-7 cells made them responsive to androgen and increased their motility in the presence to androgen. Patients with estrogen receptor positive (ER+)/AR+ BC treated with AIs were at increased risk of disease progression compared to ER+/AR+ non-AI treated and ER+/AR- AI treated cases. CONCLUSION: AIs may lead to unfavorable responses in some ER/AR positive BC cases, especially in patients with AR+, p53 mutant tumors.


Androgens/pharmacology , Aromatase Inhibitors/toxicity , Chemokine CXCL12/metabolism , Gene Expression/drug effects , Receptors, CXCR4/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CXCL12/analysis , Chemokine CXCL12/genetics , Female , Humans , MCF-7 Cells , Nuclear Receptor Coactivator 1/antagonists & inhibitors , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Promoter Regions, Genetic , RNA Interference , Receptors, Androgen/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, Estrogen/metabolism , Testosterone/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
19.
Exp Toxicol Pathol ; 69(7): 424-429, 2017 Sep 05.
Article En | MEDLINE | ID: mdl-28410883

Aminoglutethimide is a steroidogenesis inhibitor and inhibits a cholesterol side-chain cleavage enzyme (CYP11A1) that converts cholesterol to pregnenolone in mitochondria. We investigated histopathological changes induced by 5-day administration of AG in mice. Cytoplasmic vacuoles of various sizes and single cell necrosis were found in zona fasciculata cells in AG-treated mice. Some vacuoles were positive for adipophilin, whereas others were positive for lysosome-associated membrane protein-2 on immunohistochemical staining, indicating they were enlarged lipid droplets and lysosomes, respectively. Electron microscopy revealed enlarged lysosomes containing damaged mitochondria and lamellar bodies in zona fasciculata cells, and they were considered to reflect the intracellular protein degradation processes, mitophagy and lipophagy. From these results, we showed that AG induces excessive lipid accumulation and mitochondrial damage in zona fasciculata cells, which leads to an accelerated lysosomal degradation in mice.


Adrenal Glands/drug effects , Adrenal Glands/pathology , Aminoglutethimide/toxicity , Aromatase Inhibitors/toxicity , Lysosomes/drug effects , Animals , Lysosomes/pathology , Male , Mice
20.
Environ Sci Technol ; 51(8): 4661-4672, 2017 04 18.
Article En | MEDLINE | ID: mdl-28355063

A quantitative adverse outcome pathway (qAOP) consists of one or more biologically based, computational models describing key event relationships linking a molecular initiating event (MIE) to an adverse outcome. A qAOP provides quantitative, dose-response, and time-course predictions that can support regulatory decision-making. Herein we describe several facets of qAOPs, including (a) motivation for development, (b) technical considerations, (c) evaluation of confidence, and (d) potential applications. The qAOP used as an illustrative example for these points describes the linkage between inhibition of cytochrome P450 19A aromatase (the MIE) and population-level decreases in the fathead minnow (FHM; Pimephales promelas). The qAOP consists of three linked computational models for the following: (a) the hypothalamic-pitutitary-gonadal axis in female FHMs, where aromatase inhibition decreases the conversion of testosterone to 17ß-estradiol (E2), thereby reducing E2-dependent vitellogenin (VTG; egg yolk protein precursor) synthesis, (b) VTG-dependent egg development and spawning (fecundity), and (c) fecundity-dependent population trajectory. While development of the example qAOP was based on experiments with FHMs exposed to the aromatase inhibitor fadrozole, we also show how a toxic equivalence (TEQ) calculation allows use of the qAOP to predict effects of another, untested aromatase inhibitor, iprodione. While qAOP development can be resource-intensive, the quantitative predictions obtained, and TEQ-based application to multiple chemicals, may be sufficient to justify the cost for some applications in regulatory decision-making.


Aromatase Inhibitors/toxicity , Fadrozole/toxicity , Animals , Cyprinidae , Estradiol/metabolism , Models, Theoretical , Predictive Value of Tests , Vitellogenins/metabolism
...