Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Toxics ; 11(7)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37505567

ABSTRACT

The use of zinc oxide nanoparticles (ZnO NP) in consumer products is increasing, raising concern about their potential toxicity to human health. Nanoparticles have endocrine disrupting effects and can induce oxidative stress, leading to biomolecule oxidation and cell dysfunction. The ovary is one of the most important endocrine organs in female reproduction. Nanoparticles accumulate in the ovary, but it is unknown whether and how exposure to these materials disrupts antral follicle functions. Thus, this study tested the hypothesis that the in vitro exposure to ZnO NPs affects the steroidogenic pathway and induces oxidative stress in ovarian antral follicles. Antral follicles from CD-1 mice were cultured with ZnO NPs (5, 10, and 15 µg/mL) for 96 h. ZnO NP exposure did not affect apoptosis and cell cycle regulators at any of the tested concentrations. ZnO NP exposure at low levels (5 µg/mL) increased aromatase levels, leading to increased estradiol levels and decreased estrogen receptor alpha (Esr1) expression. ZnO NP exposure at 15 µg/mL induced an antioxidant response in the antral follicles as evidenced by changes in expression of antioxidant molecules (Nrf2, Cat, Sod1, Gsr, Gpx) and decreased levels of reactive oxygen species. Interestingly, ZnO NPs dissolve up to 50% in media and are internalized in cells as soon as 1 h after culture. In conclusion, ZnO NPs are internalized in antral follicles, leading to increased estrogen production and an antioxidant response.

2.
Toxicol Sci ; 184(1): 57-66, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34382084

ABSTRACT

Temephos is an organophosphorus pesticide used in control campaigns against vectors that transmit diseases, including dengue, a public health concern. The WHO classifies temephos in category III and its safe concentration (low-observable-adverse-effect level) in male rats is 100 mg/kg/day for up to 44 days. Temephos inhibits acetylcholinesterase (AChE) and is metabolized in different tissues, probably by mixed-function oxidases; one of its metabolites is bisphenol S (BPS), which is considered an endocrine disruptor. The aim of this study was to evaluate the effects of temephos on sperm function and its biotransformation in the testis, epididymis, and other tissues to explore its toxicity in rats treated with 100 mg/kg/day/5 or 7 days (gavage). AChE activity was inhibited 70% starting on day 3 and 13 or 41% mortality was observed at 5 or 7 days, respectively. After 7 days, temephos significantly decreased sperm motility (30%) and viability (10%) and increased (10%) lipoperoxidation, and the sperm DNA exhibited no damage. Temephos was distributed and metabolized in all tissues, with the highest levels observed in the adipose tissue and temephos levels were 16-fold higher in the epididymis than in the testis. Notably, BPS was observed in the testis. At 5 days, decreased sperm motility (12.5%) and viability (5.7%) were observed and sperm fertilization decreased (30%). These results suggest that temephos decreases sperm quality and fertilization capacity at recommended safe concentrations and that it is metabolized in male reproductive tissues. This pesticide places the reproductive health of exposed people at risk, suggesting the need to reevaluate its toxicity.


Subject(s)
Pesticides , Temefos , Acetylcholinesterase/metabolism , Animals , Epididymis , Fertilization , Humans , Male , Organophosphorus Compounds , Pesticides/toxicity , Rats , Sperm Motility , Spermatozoa , Temefos/toxicity , Testis
3.
Nutrients ; 13(8)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34444776

ABSTRACT

Cyclophosphamide (CP)-which is used to treat autoimmune diseases and cancer-is related to gonadotoxicity attributed to oxidative stress. As phycobiliproteins (PBPs) are strong antioxidants that are unexplored as protective agents against male gonadotoxicity, our work aimed to investigate the effects of PBP crude extract on testicular damage and sperm parameter alterations caused by CP in mice. Three doses of PBP (50, 100, and 200 mg/kg) were tested in the experimental groups (n = 8 per group), administered concomitantly with 100 mg/kg CP. After 42 days receiving PBP daily and CP weekly, body and relative testicular weights, serum testosterone levels, testicular lipoperoxidation and antioxidant enzyme activity levels, and testicular histology and sperm parameter alterations were assessed. The results showed that PBP crude extract at 200 mg/kg prevented testosterone serum reduction, body weight loss, lipoperoxidation and enzyme activity increments, and sperm parameter alterations and partially ameliorated relative testicular weight reductions and histological damage in CP-treated mice. In conclusion, we showed that PBP crude extract (200 mg/kg) mitigated oxidative damage in the testes and ameliorated alterations in sperm parameters in mice treated with CP (100 mg/kg); therefore, PBP extract could be considered as a potential protective agent against CP toxicity.


Subject(s)
Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Phycobiliproteins/toxicity , Animals , Antioxidants/pharmacology , Body Weight , Disease Models, Animal , Male , Mice , Oxidative Stress/drug effects , Protective Agents/pharmacology , Seminiferous Tubules/drug effects , Seminiferous Tubules/pathology , Spermatozoa/drug effects , Testis/drug effects , Testis/pathology , Testosterone/blood
4.
Reprod Toxicol ; 103: 79-95, 2021 08.
Article in English | MEDLINE | ID: mdl-34098047

ABSTRACT

The ovary is a highly important organ for female reproduction. The main functions include sex steroid hormone synthesis, follicular development, and achievement of oocyte meiotic and development competence for proper fertilization. Nanoparticle (NP) exposure is becoming unavoidable because of its wide use in different products, including cosmetics, food, health, and personal care products. Studies examining different nonreproductive tissues or systems have shown that characteristics such as the size, shape, core material, agglomeration, and dissolution influence the effects of NPs. However, most studies evaluating NP-mediated reproductive toxicity have paid little or no attention to the influence of the physicochemical characteristics of NP on the observed effects. As accumulating evidence indicates that NP may reach the ovary to impair proper functions, this review summarizes the available data on NP accumulation in ovarian tissue, as well as data describing toxicity to ovarian functions, including sex steroid hormone production, follicular development, oocyte quality, and fertility. Due to their toxicological relevance, this review also describes the main physicochemical characteristics involved in NP toxicity and the importance of considering NP physicochemical characteristics as factors influencing the ovarian toxicity of NPs. Finally, this review summarizes the main mechanisms of toxicity described in ovarian cells.


Subject(s)
Hazardous Substances/toxicity , Nanoparticles/toxicity , Ovary/drug effects , Animals , Female , Fertility , Humans , Oocytes , Reproduction
5.
Reprod Toxicol ; 96: 399-412, 2020 09.
Article in English | MEDLINE | ID: mdl-32798650

ABSTRACT

Titanium dioxide (TiO2) and zinc oxide (ZnO) nanoparticles (NP) have been demonstrated to reach the ovary. However, the potential detrimental effects of these metal-based NP on ovarian antral follicles and whether they can be directly taken up by follicular cells are unknown. The aim of this study was to evaluate whether TiO2 and ZnO NP internalize into the antral follicle, and further compared any potential detrimental effects of either NP on growth, ultrastructure and viability of antral follicles. It has been described that TiO2 and ZnO NP induce oxidative stress, thus this study indirectly assessed whether oxidative stress was involved. Antral follicles were cultured with TiO2 (5, 25 and 50 µg/mL) or ZnO (5, 15 and 25 µg/mL) NP for 96 h. TiO2 NP were internalized and agglomerated into cells, increased follicle diameter and disrupted the cytoskeleton arrangement, effects that were partially prevented by a co-exposure with trolox. Moreover, ZnO NP partially dissolved into culture media, decreased follicle diameter, and disrupted cytoskeletal arrangement, and these effects were not prevented by trolox. Ultrastructural alterations induced by exposure to both NP were evidenced by impaired transzonal projections and swelling mitochondria. Oxidative stress mediates TiO2 NP-induced effects but not those from ZnO NP in antral follicle development. Our results suggest that both NP induced ovarian follicle toxicity through different toxic mechanisms, possibly due to a stimulation of ZnO NP solubility and agglomeration of TiO2 NP into the follicular cells.


Subject(s)
Nanoparticles/administration & dosage , Ovarian Follicle/drug effects , Titanium/administration & dosage , Zinc Oxide/administration & dosage , Animals , Cytoskeleton/drug effects , Female , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovarian Follicle/ultrastructure , Oxidative Stress/drug effects
6.
Nutr Metab Cardiovasc Dis ; 30(1): 40-48, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31757567

ABSTRACT

BACKGROUND AND AIMS: Paraoxonase 1 (PON1) is considered to play a crucial role as an anti-atherosclerotic factor. The PON1 activity is affected by genetic polymorphisms, environmental factors, age, sex, lifestyle, pharmaceutical drugs, and dietary factors. The aim of this study was to evaluate the association between macro- and micronutrients as well as PON1 concentration and activities in patients with cardiovascular diseases (CVD), cardiovascular risk factors but no CVD (CRF), and in healthy controls (control group). METHODS AND RESULTS: A case-control study was carried out with 356 volunteers from the Mexican Institute of Social Security, Mexico. Clinical parameters, lipid profile, PON1 activities (AREase, LACase, CMPAase and PONase), and PON1 concentration were evaluated. There was a differential intake of macro- and micronutrients among the study groups. The intake of proteins and carbohydrates was higher in the CVD group than in the CFR and control groups (p < 0.05). AREase, LACase, and CMPAase activities and PON1 concentration were lowest in the CVD group. CONCLUSION: LACase and CMPAase activities, as well as PON1 concentration, could be included in the battery of CVD predictive biomarkers in the Mexican population.


Subject(s)
Aryldialkylphosphatase/blood , Cardiovascular Diseases/blood , Diet , Nutritional Status , Nutritive Value , Aged , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/physiopathology , Case-Control Studies , Diet/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Energy Intake , Female , Humans , Male , Mexico/epidemiology , Micronutrients/administration & dosage , Middle Aged , Phenotype , Prognosis , Protective Factors , Risk Factors
7.
Reprod Toxicol ; 90: 53-61, 2019 12.
Article in English | MEDLINE | ID: mdl-31442482

ABSTRACT

Because di(2-ethylhexyl) phthalate (DEHP) toxicity on ovarian function is incomplete, effects of DEHP oocyte fertilization and the resulting zygotes were investigated. Further, an analysis characterizing the stage of zygote arrest was performed. Female CD1 mice were dosed orally with DEHP (0, 20, 200 and 2000 µg/kg/day) for 30 days. Following an in vivo mating post-dosing, DEHP-treated females exhibited fewer oocytes/zygotes, fewer oocytes displaying the polar body extrusion, fewer 1-cell zygotes having 2-pronuclei, more unfertilized oocytes, and decreased number of zygotes at every stage of development. DEHP induced blastomere fragmentation in zygotes. DNA replication in zygotes directly assessed by the 5-Ethynyl-2'-deoxyuridine (5-EdU) incorporation assay and indirectly by dosing mice with 5-fluorouracil (5-FU) suggested that DEHP inhibits DNA replication. Our data suggest that DEHP at doses found in 'every-day' (200 µg/Kg/day) or occupational (2000 µg/Kg/day) environments induces zygote fragmentation and arrests its development from the 2-cell stage potentially impairing DNA replication.


Subject(s)
Diethylhexyl Phthalate/toxicity , Environmental Pollutants/toxicity , Oocytes/drug effects , Plasticizers/toxicity , Zygote/drug effects , Administration, Oral , Animals , Female , Fertilization/drug effects , Male , Mice
8.
Toxicol Appl Pharmacol ; 380: 114694, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31356930

ABSTRACT

Lead (Pb) exposure at high concentrations is associated with poor sperm quality, acrosome alterations, and low fertilization rate. Sperm capacitation and the acrosome reaction (AR) are required for successful fertilization. Actin polymerization is crucial for correct capacitation, and small GTPases, such as RhoA, Rac1, and Cdc42, are involved. This study aimed to evaluate the effects of Pb on sperm fertilization ability, capacitation, AR, and the mechanisms involved in mice exposed to low Pb concentrations. CD1 mice were exposed to 0.01% Pb2+ for 45 days through their drinking water and their spermatozoa were collected from the cauda epididymis-vas deferens to evaluate the following: AR (oAR: initial, sAR: spontaneous, and iAR: induced) using the PNA-FITC assay, sperm capacitation (P-Tyr levels), actin polymerization (phalloidin-TRITC), MDA production (stress oxidative marker), the RhoA, Rac1, and Cdc42 protein levels, and the in vitro fertilization (IVF). After the treatment, the blood Pb (PbB) concentration was 9.4 ±â€¯1.6 µg/dL. Abnormal sperm morphology and the oAR increased (8 and 19%, respectively), whereas the iAR decreased (15%) after a calcium ionophore challenge, and the actin polymerization decreased in the sperm heads (59%) and tails (42%). Rac1 was the only Rho protein to significantly decrease (33%). Spermatozoa from the Pb-treated mice showed a significant reduction in the fertilization rate (19%). Our data suggest that Pb exposure at environmental concentrations (PbB < 10 µg/dL) decreases the acrosome function and affects the sperm fertilization ability; this is probably a consequence of the low Rac1 levels, which did not allow adequate actin polymerization to occur.


Subject(s)
Environmental Pollutants/toxicity , Lead/toxicity , Sperm Capacitation/drug effects , Spermatozoa/drug effects , Actins/metabolism , Animals , Female , Male , Mice, Inbred ICR , Neuropeptides/metabolism , Spermatozoa/abnormalities , Spermatozoa/physiology , rac1 GTP-Binding Protein/metabolism
9.
Reprod Toxicol ; 86: 86-97, 2019 06.
Article in English | MEDLINE | ID: mdl-31028817

ABSTRACT

The aim was to evaluate the effect of perinatal BPA exposure of one or both parents on the implantation index and expression of talin, occludin and E-cadherin in the uterine epithelial cells (UEC) of the offspring. Pregnant Wistar dams (F0) received BPA or vehicle from gestational day (GD) 6 to lactation day 21. F1 animals were mated forming four groups: Control dam-Control sire (C♀-C♂), BPA dam -Control sire (B♀-C♂), Control dam -BPA sire (C♀-B♂), BPA dam -BPA sire (B♀-B♂). F1 dams were sacrificed at GD 6. Significantly decreased number of implantation sites was observed in the B♀-B♂ group as compared to the C♀-C♂ group, which correlated with decreased talin apical/basal expression ratio, occludin apical expression, and E-cadherin apical/lateral expression ratio in the UEC. Furthermore, decreased E-cadherin expression in the blastocyst was observed. Our data suggest that reduced protein expressions in F1 BPA offspring could result from decreased progesterone serum levels.


Subject(s)
Benzhydryl Compounds/toxicity , Cadherins/metabolism , Embryo Implantation/drug effects , Maternal-Fetal Exchange , Occludin/metabolism , Phenols/toxicity , Talin/metabolism , Animals , Estradiol/blood , Female , Male , Pregnancy , Progesterone/blood , Rats, Wistar , Uterus/drug effects , Uterus/metabolism
10.
Toxicol Appl Pharmacol ; 344: 13-22, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29458137

ABSTRACT

In ovarian follicles, cumulus cells communicate with the oocyte through gap junction intercellular communication (GJIC), to nurture the oocyte and control its meiosis arrest and division. Bisphenol A (BPA) is a monomer found in polycarbonate-made containers that can induce functional alterations, including impaired oocyte meiotic division and reduced molecule transfer in GJIC. However, how BPA alters oocyte meiotic division is unclear. We investigated whether BPA effects on oocyte meiotic division were correlated with reduced transfer in GJIC. Cumulus cell-oocyte complexes (COCs) isolated from mouse preovulatory follicles were cultured with 0, 0.22, 2.2, 22, 220, and 2200 nM BPA for 2 h. An additional 16-h incubation with epidermal growth factor (EGF) was performed to promote the occurrence of meiotic resumption and progression to metaphase II. Without EGF stimulus, BPA treatment increased the percentage of oocytes undergoing meiotic resumption, decreased GJIC in the COCs, and did not modify GJIC gene (Cx43 and Cx37) and protein (CX43) expression. Following EGF stimulus, BPA increased the percentage of oocytes that remained at the anaphase and telophase stages, and decreased the percentage of oocytes reaching the metaphase II stage. Concomitantly, BPA reduced the expansion of cumulus cells. Carbenoxolone (a GJIC inhibitor) and 6-diazo-5-oxo-l-norleucine (a cumulus cell-expansion inhibitor) exerted effects on meiotic division similar to those exerted by BPA. These data suggest that BPA accelerates meiotic progression, leading to impaired prophase I-to-metaphase II transition, and that this adverse effect is correlated with reduced bidirectional communication in the COC.


Subject(s)
Benzhydryl Compounds/toxicity , Cumulus Cells/physiology , Estrogens, Non-Steroidal/toxicity , Gap Junctions/physiology , Oocytes/physiology , Oogenesis/physiology , Phenols/toxicity , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cumulus Cells/drug effects , Dose-Response Relationship, Drug , Female , Gap Junctions/drug effects , Meiosis/drug effects , Meiosis/physiology , Mice , Mice, Inbred C57BL , Oocytes/drug effects , Oogenesis/drug effects
11.
Environ Toxicol ; 32(6): 1754-1764, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28233943

ABSTRACT

The indiscriminate use of pesticides in agriculture and public health campaigns has been associated with an increase of oxidative stress and DNA damage, resulting in health outcomes. Some defense mechanisms against free radical-induced oxidative damage include the antioxidant enzyme systems. The aim of this study was to determine the levels of malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), and the relationship of antioxidant enzyme levels with DNA damage among sprayers (workers) occupationally exposed to pesticides. The determinations of MDA and antioxidant enzymes were performed spectrophotometrically. The genotoxic effects were evaluated using the comet assay. The results showed a marginally significant decrease in SOD and CAT activities in the high exposure group compared to the control group. For MDA, statistically significant differences were found among people working long term vs. those working temporarily (P = 0.02) as sprayers. In the moderate exposure group, a positive correlation was observed between MDA levels and GPx activity. In the high exposure group, a negative correlation was observed between GR and CAT activities, and between MDA levels and GPx activities. Furthermore, in the high exposure group, a positive correlation between DNA damage parameters and MDA levels was observed. The results suggest an important role of antioxidant enzymes for the protection of DNA damage caused by occupational exposure to pesticides.


Subject(s)
DNA Damage , Occupational Exposure/adverse effects , Organophosphates/toxicity , Oxidative Stress/drug effects , Pesticides/toxicity , Pyrethrins/toxicity , Antioxidants/metabolism , Catalase/blood , Comet Assay , Cross-Sectional Studies , Glutathione Peroxidase/blood , Glutathione Reductase/blood , Humans , Malondialdehyde/blood , Occupational Exposure/analysis , Superoxide Dismutase/blood
12.
Toxicol Appl Pharmacol ; 289(3): 507-14, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26493930

ABSTRACT

Follicle growth culminates in ovulation, which allows for the expulsion of fertilizable oocytes and the formation of corpora lutea. Bisphenol A (BPA) is present in many consumer products, and it has been suggested that BPA impairs ovulation; however, the underlying mechanisms are unknown. Therefore, this study first evaluated whether BPA alters ovulation by affecting folliculogenesis, the number of corpora lutea or eggs shed to the oviduct, ovarian gonadotropin responsiveness, hormone levels, and estrous cyclicity. Because it has been suggested (but not directly confirmed) that BPA exerts toxic effects on the fertilization ability of oocytes, a second aim was to evaluate whether BPA impacts the oocyte fertilization rate using an in vitro fertilization assay and mating. The possible effects on early zygote development were also examined. Young adult female C57BL/6J mice (39 days old) were orally dosed with corn oil (vehicle) or 50 µg/kgbw/day BPA for a period encompassing the first three reproductive cycles (12-15 days). BPA exposure did not alter any parameters related to ovulation. Moreover, BPA exposure reduced the percentage of fertilized oocytes after either in vitro fertilization or mating, but it did not alter the zygotic stages. The data indicate that exposure to the reference dose of BPA does not impact ovulation but that it does influence the oocyte quality in terms of its fertilization ability.


Subject(s)
Benzhydryl Compounds/adverse effects , Fertilization/drug effects , Oocytes/drug effects , Ovulation/drug effects , Phenols/adverse effects , Animals , Estrous Cycle/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Ovarian Follicle/drug effects , Zygote/drug effects
13.
Toxicol Appl Pharmacol ; 279(3): 391-400, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24998973

ABSTRACT

Methamidophos (MET) is a highly toxic organophosphate (OP) pesticide that is widely used in developing countries. MET has male reproductive effects, including decreased fertility. We evaluated MET effects on sperm quality, fertilization and DNA integrity, exploring the sensitivity of different stages of spermatogenesis. Adult male mice received MET (3.75 or 5mg/kg-bw/ip/day/4 days) and were euthanized 1, 28 or 45 days post-treatment (dpt) to evaluate MET's effects on epididymal maturation, meiosis or mitosis, respectively. Spermatozoa were obtained from the cauda epididymis-vas deferens and were evaluated for sperm quality, acrosome reaction (AR; Coomassie staining), mitochondrial membrane potential (by JC-1), DNA damage (comet assay), oxidative damage (malondialdehyde (MDA) production), in vitro fertilization and protein phosphorylation (immunodetection), and erythrocyte acetylcholinesterase (AChE) activity. At 1-dpt, MET inhibited AChE (43-57%) and increased abnormal cells (6%). While at 28- and 45-dpt, sperm motility and viability were significantly reduced with an increasing MET dose, and abnormal morphology increased at 5mg/kg/day/4 days. MDA and mitochondrial activity were not affected at any dose or time. DNA damage (OTM and %DNA) was observed at 5mg/kg/day/4 days in a time-dependent manner, whereas both parameters were altered in cells from mice exposed to 3.75 mg/kg/day/4 days only at 28-dpt. Depending on the time of collection, initial-, spontaneous- and induced-AR were altered at 5mg/kg/day/4 days, and the fertilization capacity also decreased. Sperm phosphorylation (at serine and tyrosine residues) was observed at all time points. Data suggest that meiosis and mitosis are the more sensitive stages of spermatogenesis for MET reproductive toxicity compared to epididymal maturation.


Subject(s)
DNA Replication/drug effects , Insecticides/toxicity , Organothiophosphorus Compounds/toxicity , Spermatogenesis/drug effects , Spermatozoa/drug effects , Acetylcholinesterase/metabolism , Acrosome Reaction/drug effects , Animals , Body Weight/drug effects , Comet Assay , Female , Fertilization/drug effects , In Vitro Techniques , Infertility, Male/chemically induced , Infertility, Male/pathology , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred ICR , Oocytes/drug effects , Organ Size/drug effects , Oxidative Stress/drug effects , Phosphorylation , Reproduction/drug effects
14.
Environ Toxicol ; 29(8): 942-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23125138

ABSTRACT

Methamidophos (MET), widely used in developing countries, is a highly neurotoxic organophosphate pesticide that has been associated with male reproductive alterations. Commercial formulations of pesticides used by agricultural workers and urban sprayers are responsible for thousands of intoxications in developing countries and may not have the same effects as active pure ingredients. Therefore, we compared effects of MET technical (METt) and commercial (METc) grades on sperm quality and DNA integrity. Male mice were injected (intraperitoneal, i.p.) with METt or METc (3.75, 5, and 7 mg/kg bw/day/4 days) and sacrificed 24 h post-treatment. Sperm cells collected from epididymis-vas deferens were evaluated for quality parameters, DNA damage by the comet assay, and lipoperoxidation by malondialdehyde (MDA) production. Erythrocyte acetylcholinesterase (AChE) activity was evaluated by acetylthiocholine inhibition as an index of overall toxicity. A dose-dependent AChE inhibition was observed with both formulations. Sperm quality was decreased after treatment with both MET compounds, but the commercial formulation showed stronger effects; a similar profile was observed with the DNA damage, being METc more genotoxic. None MET formulation increased MDA, suggesting no peroxidative damage involved. In summary, the commercial formulation of MET was more reprotoxic and genotoxic than the active pure ingredient, highlighting that commercial formulations must be considered for more appropriate risk assessment of pesticide exposures.


Subject(s)
DNA Damage , Organothiophosphorus Compounds/toxicity , Pesticides/toxicity , Acetylcholinesterase/blood , Animals , Comet Assay , DNA/pharmacology , Erythrocytes/drug effects , Erythrocytes/enzymology , Lipid Peroxidation , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred ICR , Reproduction/drug effects , Spermatozoa/cytology , Spermatozoa/drug effects
15.
Reprod Biol Endocrinol ; 11: 26, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23548098

ABSTRACT

BACKGROUND: Previous studies have demonstrated that pre-pubertal aryl hydrocarbon receptor knockout (AHRKO) mice have slow antral follicle growth and reduced capacity to produce estradiol compared to wild-type (WT) mice. Although previous studies have suggested that this is likely due to a reduced ability of the AHRKO follicles to respond to follicle-stimulating hormone (FSH), this possibility was not directly tested. Thus, the goal of these studies was to test the hypothesis that low FSH responsiveness is responsible for the slow growth and reduced estradiol production observed in pre-pubertal AHRKO versus WT antral follicles. METHODS: Antral follicles from WT and AHRKO mice were cultured with varying amounts of FSH (0-15 IU/mL) for up to 7 days, and subjected to measurements of growth, FSH receptor and steroidogenic regulator expression, sex steroid hormone levels, and inhibin beta-A expression. General linear models (GLM) for repeated measures were used to compare follicle diameters over time among treatments. If the global tests from GLM were significant, Tukey's tests were used for pairwise comparisons. Remaining comparisons among groups were performed using one-way analysis of variance followed by Tukey's post hoc test. RESULTS: The results indicate that FSH stimulated growth in both WT and AHRKO follicles, but that high levels of FSH (10-15 IU/mL) were required for AHRKO follicles to reach maximal growth, whereas lower levels of FSH (5 IU/mL) were required for WT follicles to reach maximal growth. Further, FSH stimulated expression of FSH receptor, steroidogenic factors, and inhibin beta-A as well as production of steroid hormones in both WT and AHRKO follicles, but the degree of stimulation differed between WT and AHRKO follicles. Interestingly, FSH treatment increased expression of FSH receptor, some steroidogenic regulators, inhibin beta-A, and steroid hormone production more in AHRKO follicles compared to WT follicles. CONCLUSIONS: Collectively, these data suggest that the slow growth, but not reduced steroidogenesis in AHRKO follicles, is due to their reduced ability to respond to FSH compared to WT follicles. These data also suggest that the AHR may contribute to the ability of FSH to stimulate proper follicle growth, but it may not contribute to FSH-induced steroidogenesis.


Subject(s)
Estradiol/metabolism , Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/drug effects , Receptors, Aryl Hydrocarbon/deficiency , Animals , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Inhibin-beta Subunits/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Phosphoproteins/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, FSH/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Culture Techniques
16.
Reprod Toxicol ; 34(1): 16-21, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22484361

ABSTRACT

Methoxychlor (MXC) is an organochlorine pesticide used against pests that attack crops, vegetables, and livestock. MXC inhibits growth and induces atresia (death) of mouse ovarian antral follicles in vitro. Since several studies indicate that many chemicals act through the aryl hydrocarbon receptor (AHR) pathway, the current study tested the hypothesis that MXC binds to the AHR to inhibit growth and induce atresia of antral follicles. The data indicate that MXC binds to AHR. Further, a relatively high dose of MXC (100µg/ml) inhibits growth and induces atresia in both wild-type (WT) and AHR null (AHRKO) follicles, whereas a lower dose of MXC (10µg/ml) inhibits growth and induces atresia in WT, but not in AHRKO follicles. These data indicate that AHR deletion partially protects antral follicles from MXC induced slow growth and atresia. Collectively, these data show that MXC may act through the AHR pathway to inhibit follicle growth and induce atresia in antral follicles of the ovary.


Subject(s)
Follicular Atresia/drug effects , Insecticides/toxicity , Methoxychlor/toxicity , Ovarian Follicle/drug effects , Receptors, Aryl Hydrocarbon/physiology , Animals , Estradiol/metabolism , Female , Follicular Atresia/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Follicle/growth & development , Ovarian Follicle/pathology
17.
Toxicol Appl Pharmacol ; 253(3): 161-9, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21514315

ABSTRACT

The organochlorine pesticide methoxychlor (MXC) is a known endocrine disruptor that affects adult rodent females by causing reduced fertility, persistent estrus, and ovarian atrophy. Since MXC is also known to target antral follicles, the major producer of sex steroids in the ovary, the present study was designed to test the hypothesis that MXC decreases estradiol (E2) levels by altering steroidogenic and metabolic enzymes in the antral follicles. To test this hypothesis, antral follicles were isolated from CD-1 mouse ovaries and cultured with either dimethylsulfoxide (DMSO) or MXC. Follicle growth was measured every 24 h for 96 h. In addition, sex steroid hormone levels were measured using enzyme-linked immunosorbent assays (ELISA) and mRNA expression levels of steroidogenic enzymes as well as the E2 metabolic enzyme Cyp1b1 were measured using qPCR. The results indicate that MXC decreased E2, testosterone, androstenedione, and progesterone (P4) levels compared to DMSO. In addition, MXC decreased expression of aromatase (Cyp19a1), 17ß-hydroxysteroid dehydrogenase 1 (Hsd17b1), 17α-hydroxylase/17,20-lyase (Cyp17a1), 3ß hydroxysteroid dehydrogenase 1 (Hsd3b1), cholesterol side-chain cleavage (Cyp11a1), steroid acute regulatory protein (Star), and increased expression of Cyp1b1 enzyme levels. Thus, these data suggest that MXC decreases steroidogenic enzyme levels, increases metabolic enzyme expression and this in turn leads to decreased sex steroid hormone levels.


Subject(s)
Estradiol/analysis , Gonadal Steroid Hormones/biosynthesis , Insecticides/toxicity , Methoxychlor/toxicity , Ovarian Follicle/drug effects , 17-Hydroxysteroid Dehydrogenases/analysis , Animals , Aromatase/analysis , Cells, Cultured , Estradiol/metabolism , Female , Mice , Ovarian Follicle/chemistry , Ovarian Follicle/metabolism , Phosphoproteins/physiology
18.
Toxicol Sci ; 120(2): 447-59, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252393

ABSTRACT

Methoxychlor (MXC), an organochlorine pesticide, and its metabolites, mono-hydroxy MXC (MOH) and bis-hydroxy MXC (HPTE) are known ovarian toxicants and can cause inhibition of antral follicle growth. Since these chemicals bind to estrogen receptor alpha (ESR1), we hypothesized that ovaries overexpressing ESR1 (ESR1 OE) would be more susceptible to toxicity induced by MXC and its metabolites because the chemicals can bind to more ESR1 in the antral follicles. We cultured antral follicles from controls and ESR1 OE mouse ovaries with either the vehicle dimethylsulfoxide (DMSO), MXC, MOH, or HPTE. The data show that at 96 h, the cultured antral follicles from ESR1 OE antral follicles are more susceptible to toxicity induced by MXC, MOH, and HPTE because low doses of these chemicals cause follicle growth inhibition in ESR1 OE mice but not in control mice. On comparing gene expression levels of nuclear receptors in the cultured antral follicles of ESR1 OE and control follicles, we found differential messenger RNA (mRNA) expression of Esr1, estrogen receptor beta (Esr2), androgen receptor (Ar), progesterone receptor (Pr), and aryl hydrocarbon receptor (Ahr) between the genotypes. We also analyzed mRNA levels of Cyp3a41a, the enzyme metabolizing MOH and HPTE, in the cultured follicles and found that Cyp3a41a was significantly lower in DMSO-treated ESR1 OE follicles compared with controls. In ESR1 OE livers, we found that Cyp3a41a levels were significantly lower compared with control livers. Collectively, these data suggest that MXC and its metabolites cause differential gene expression in ESR1 OE mice compared with controls. The results also suggest that the increased sensitivity of ESR1 OE mouse ovaries to toxicity induced by MXC and its metabolites is due to low clearance of the metabolites by the liver and ovary.


Subject(s)
Estrogen Receptor alpha/physiology , Methoxychlor/analogs & derivatives , Ovarian Follicle/drug effects , Phenols/toxicity , Animals , Cell Size/drug effects , Cells, Cultured , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 2 , Estrogen Receptor alpha/genetics , Female , Gene Expression/drug effects , Immunohistochemistry , Methoxychlor/metabolism , Methoxychlor/toxicity , Mice , Mice, Transgenic , Ovarian Follicle/enzymology , Ovarian Follicle/metabolism , Phenols/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
J Androl ; 32(3): 282-94, 2011.
Article in English | MEDLINE | ID: mdl-20930192

ABSTRACT

Previously, we reported that estrogen receptor α mRNA (Esr1) or protein (ESR1) overexpression resulting from neonatal exposure to estrogens in rats was associated with infertility and maldeveloped penis characterized by reduced length and weight and abnormal accumulation of fat cells. The objective of this study was to determine if mutant male mice overexpressing Esr1 are naturally infertile or have reduced fertility and/or develop abnormal penis. The fertility parameters, including fertility and fecundity indices, numbers of days from the day of cohabitation to the day of delivery, and numbers of pups per female, were not altered from controls as a result of Esr1 overexpression. Likewise, penile morphology, including the length, weight, and diameter and os penis development, was not altered from controls. Conversely, weights of the seminal vesicles and bulbospongiosus and levator ani (BS/LA) muscles were significantly (P < .05) lower as compared with controls; however, the weight of the testis, the morphology of the testis and epididymis, and the plasma and testicular testosterone concentration were not different from controls. Hence, genetically induced Esr1 overexpression alone, without an exogenous estrogen exposure during the neonatal period, is unable to adversely affect the development of the penis as well as other male reproductive organs, except for limited, but significant, reductions in weights of the seminal vesicles and BS/LA muscles.


Subject(s)
Estrogen Receptor alpha/genetics , Fertility/genetics , Penis/embryology , RNA, Messenger/genetics , Animals , Base Sequence , Blotting, Western , Body Weight , DNA Primers , Female , Male , Mice , Mice, Transgenic , Organ Size , Polymerase Chain Reaction , Rats , Testosterone/blood , Testosterone/metabolism
20.
Toxicol Sci ; 119(1): 209-17, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20956811

ABSTRACT

Bisphenol A (BPA) is used as the backbone for plastics and epoxy resins, including various food and beverage containers. BPA has also been detected in 95% of random urine samples and ovarian follicular fluid of adult women. Few studies have investigated the effects of BPA on antral follicles, the main producers of sex steroid hormones and the only follicles capable of ovulation. Thus, this study tested the hypothesis that postnatal BPA exposure inhibits antral follicle growth and steroidogenesis. To test this hypothesis, antral follicles isolated from 32-day-old FVB mice were cultured with vehicle control (dimethyl sulfoxide [DMSO]), BPA (4.4-440 µM), pregnenolone (10 µg/ml), pregnenolone + BPA 44 µM, and pregnenolone + BPA 440 µM. During the culture, follicles were measured for growth daily. After the culture, media was subjected to ELISA for hormones in the estradiol biosynthesis pathway, and follicles were processed for quantitative real-time PCR of steroidogenic enzymes. The results indicate that BPA (440 µM) inhibits follicle growth and that pregnenolone cotreatment was unable to restore/maintain growth. Furthermore, BPA 44 and 440 µM inhibit progesterone, dehydroepiandrosterone, androstenedione, estrone, testosterone, and estradiol production. Pregnenolone cotreatment was able to increase production of pregnenolone, progesterone, and dehydroepiandrosterone and maintain androstenedione and estrone levels in BPA-treated follicles compared with DMSO controls but was unable to protect testosterone or estradiol levels. Furthermore, pregnenolone was unable to protect follicles from BPA-(44-440 µM) induced inhibition of steroidogenic enzymes compared with the DMSO control. Collectively, these data show that BPA targets the estradiol biosynthesis pathway in the ovary.


Subject(s)
Environmental Pollutants/toxicity , Gonadal Steroid Hormones/antagonists & inhibitors , Ovarian Follicle , Phenols/toxicity , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Benzhydryl Compounds , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Estradiol/biosynthesis , Female , Gene Expression/drug effects , Gonadal Steroid Hormones/biosynthesis , In Vitro Techniques , Mice , Mice, Inbred Strains , Ovarian Follicle/drug effects , Ovarian Follicle/enzymology , Ovarian Follicle/growth & development , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Pregnenolone/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...