Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 96(5): 1105-1117, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28486587

ABSTRACT

Dibutyl phthalate (DBP) is present in consumer products and the coating of some oral medications. Acetyl tributyl citrate (ATBC) has been proposed as an alternative to DBP because DBP causes endocrine disruption in animal models. Following ingestion, DBP is converted to its main metabolite mono-butyl phthalate (MBP) which has been detected in >90% of human follicular fluid samples. Previous studies show that DBP reduces the number of antral follicles present in the ovaries of mice. Thus, this study was designed to evaluate the effects of DBP, MBP, and ATBC on in vitro growth and viability of mouse ovarian antral follicles. Antral follicles were isolated from CD-1 females (PND32-37) and treated with vehicle, DBP, MBP, or ATBC (starting at 0.001 and up to 1000 µg/ml for DBP; 24-72 h). Follicle diameter, ATP production, qPCR, and TUNEL were used to measure follicle growth, viability, cell cycle and apoptosis gene expression, and cell death-associated DNA fragmentation, respectively. While MBP did not cause toxicity, DBP exposure at ≥10 µg/ml resulted in growth inhibition followed by cytoxicity at ≥500 µg/ml. ATBC increased the number of nongrowing follicles at 0.01 µg/ml and did not affect ATP production, but increased TUNEL positive area in treated follicles. Gene expression results suggest that cytotoxicity in DBP-treated follicles occurs via activation of cell cycle arrest prior to follicular death. These findings suggest that concentrations of DBP ≥10 µg/ml are detrimental to antral follicles and that ATBC should be examined further as it may disrupt antral follicle function at low concentrations.


Subject(s)
Citrates/toxicity , Dibutyl Phthalate/toxicity , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Plasticizers/toxicity , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis Regulatory Proteins/biosynthesis , Cell Cycle/drug effects , Cell Death/drug effects , DNA Fragmentation/drug effects , Female , Gene Expression/drug effects , Mice
2.
J Appl Toxicol ; 37(6): 668-675, 2017 06.
Article in English | MEDLINE | ID: mdl-27866379

ABSTRACT

Acetyl tributyl citrate (ATBC), is a phthalate substitute used in food and medical plastics, cosmetics and toys. Although systemically safe up to 1000 mg kg-1 day-1 , its ability to cause reproductive toxicity in females at levels below 50 mg kg-1 day-1 has not been examined. This study evaluated the effects of lower ATBC exposures on female reproduction using mice. Adult CD-1 females (n = 7-8 per treatment) were dosed orally with tocopherol-stripped corn oil (vehicle), 5 or 10 mg kg-1 day-1 ATBC daily for 15 days, and then bred with a proven breeder male. ATBC exposure did not alter body weights, estrous cyclicity, and gestational and litter parameters. Relative spleen weight was slightly increased in the 5 mg kg-1 day-1 group. ATBC at 10 mg kg-1 day-1 targeted ovarian follicles and decreased the number of primordial, primary and secondary follicles present in the ovary. These findings suggest that low levels of ATBC may be detrimental to ovarian function, thus, more information is needed to understand better the impact of ATBC on female reproduction. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Citrates/toxicity , Maternal Exposure/adverse effects , Ovary/drug effects , Plasticizers/toxicity , Reproduction/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Female , Mice, Inbred Strains , Organ Size/drug effects , Ovary/pathology
3.
Reprod Toxicol ; 53: 15-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25765776

ABSTRACT

Di-n-butyl phthalate (DBP) is present in many beauty and medical products. Human exposure estimates range from 0.007-0.01 mg/kg/day in the general population and up to 0.233 mg/kg/day in patients taking DBP-coated medications. Levels of phthalates tend to be higher in women, thus, evaluating ovarian effects of DBP exposure is of great importance. Mice were given corn oil (vehicle) or DBP at 0.01, 0.1, and 1000 mg/kg/day (high dose) for 10 days to test whether DBP causes ovarian toxicity. Estrous cyclicity, steroidogenesis, ovarian morphology, and apoptosis and steroidogenesis gene expression were evaluated. DBP exposure decreased serum E2 at all doses, while 0.1DBP increased FSH, decreased antral follicle numbers, and increased mRNA encoding pro-apoptotic genes (Bax, Bad, Bid). Interestingly, mRNAs encoding the steroidogenic enzymes Hsd17b1, Cyp17a1 and Cyp19a1 were increased in all DBP-treated groups. These novel findings show that DBP can disrupt ovarian function in mice at doses relevant to humans.


Subject(s)
Dibutyl Phthalate/toxicity , Ovary/drug effects , Plasticizers/toxicity , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Cytochrome P-450 Enzyme System/genetics , Estradiol/blood , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation/drug effects , Luteinizing Hormone/blood , Mice , Ovary/pathology , Phosphoproteins/genetics
4.
Comp Med ; 63(1): 6-12, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23561932

ABSTRACT

Ovarian cancer has a high mortality rate because there are few symptoms in early disease development. The incidence of ovarian cancer increases in women after menopause. Understanding early events in this disease can best be achieved by using animal models. Therefore, the objective of this study was to develop and track the onset of ovarian tumorigenesis in mice mimicking characteristics of postmenopausal epithelial cancer in women. Female B6C3F1 mice (age, 28 d) received 4-vinylcyclohexene diepoxide (VCD, 160 mg/kg IV daily for 20 d) to cause ovarian failure. Four months after VCD treatment, via surgical intervention, each mouse received a single injection of 7,12-dimethylbenz[a]anthracene (DMBA) or vehicle control (sesame oil) under the bursa of the right ovary to cause ovarian neoplasms. The experimental groups were untreated controls (Con-Con), DMBA-treatment only (Con-DMBA), VCD treatment only (VCD-Con), and VCD+DMBA-treated (VCD+DMBA) mice. At 3, 5, 7, and 9 mo after DMBA injection, ovaries were collected for histologic and immunohistochemical evaluation. No tumors developed in Con-Con mice. All VCD-treated mice (with or without DMBA) exhibited ovarian failure. Mice that received both VCD and DMBA exhibited tumors at 3 mo (50%), 5 mo (14%), 7 mo (90%), and 9 mo (57%) after DMBA treatment; 31% of the tumors were epithelial in origin. Our findings confirm that inducing ovarian tumors in mice by chemical means is an effective method for studying early stages of tumor development that may be relevant to epithelial ovarian cancers that arise in postmenopausal women.


Subject(s)
Benz(a)Anthracenes/toxicity , Disease Models, Animal , Ovarian Neoplasms/chemically induced , Ovary/pathology , Postmenopause , Animals , Cyclohexenes , Female , Immunohistochemistry , Mice , Ovarian Neoplasms/pathology , Time Factors , Vinyl Compounds
5.
Toxicol Appl Pharmacol ; 267(1): 49-56, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23274565

ABSTRACT

4-Vinylcyclohexene diepoxide (VCD) destroys ovarian primordial and small primary follicles via apoptosis. In mice, VCD exposure induces ovarian mRNA expression of glutathione S-transferase (GST) family members, including isoform mu (Gstm). Extra-ovarian GSTM negatively regulates pro-apoptotic apoptosis signal-regulating kinase 1 (ASK1) through protein complex formation, which dissociates during stress, thereby initiating ASK1-induced apoptosis. The present study investigated the ovarian response of Gstm mRNA and protein to VCD. Induction of Ask1 mRNA at VCD-induced follicle loss onset was determined. Ovarian GSTM:ASK1 protein complex formation was investigated and VCD exposure effects thereon evaluated. Phosphatidylinositol-3 kinase (PI3K) regulation of GSTM protein was also studied. Postnatal day (PND) 4 rat ovaries were cultured in control media ± 1) VCD (30 µM) for 2-8 days; 2) VCD (30 µM) for 2 days, followed by incubation in control media for 4 days (acute VCD exposure); or 3) LY294002 (20 µM) for 6 days. VCD exposure did not alter Gstm mRNA expression, however, GSTM protein increased (P<0.05) after 6 days of both the acute and chronic treatments. Ask1 mRNA increased (0.33-fold; P<0.05) relative to control after 6 days of VCD exposure. Ovarian GSTM:ASK1 protein complex formation was confirmed and, relative to control, the amount of GSTM bound to ASK1 increased 33% (P<0.05) by chronic but with no effect of acute VCD exposure. PI3K inhibition increased (P<0.05) GSTM protein by 40% and 71% on d4 and d6, respectively. These findings support involvement of GSTM in the ovarian response to VCD exposure, through regulation of pro-apoptotic ASK1.


Subject(s)
Cyclohexenes/toxicity , Glutathione Transferase/physiology , MAP Kinase Kinase Kinase 5/metabolism , Ovary/drug effects , Ovary/metabolism , Vinyl Compounds/toxicity , Animals , Animals, Newborn , Cells, Cultured , Female , Ovary/enzymology , Rats , Rats, Inbred F344
6.
Toxicol Appl Pharmacol ; 258(1): 118-23, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22061827

ABSTRACT

4-vinylcyclohexene diepoxide (VCD) is a metabolite of 4-vinylcyclohexene (VCH) which has the potential to be formed in the ovary through CYP2E1 activity. VCD specifically destroys primordial and small primary follicles in the rodent ovary. Mouse ovaries exposed to VCD demonstrate increased mRNA and protein expression of microsomal epoxide hydrolase (mEH), and an inactive tetrol metabolite (4-(1,2-dihydroxy)ethyl-1,2-dihydroxycyclohexane) can be formed in mouse ovarian follicles, potentially through detoxification action of mEH. In contrast, mEH can bioactivate another ovotoxic chemical, 7,12-dimethylbenz[a]anthracene (DMBA) to a more toxic compound, DMBA-3,4-diol-1,2-epoxide. Thus, the present study evaluated a functional role for mEH during detoxification of VCD. Additionally, because inhibition of the phosphatidyinositol-3 kinase (PI3K) signaling pathway in a previous study protected primordial follicles from VCD-induced destruction, but accelerated DMBA-induced ovotoxicity, a role for PI3K in ovarian mEH regulation was evaluated. Using a post-natal day (PND) 4 Fischer 344 rat whole ovary culture system inhibition of mEH using cyclohexene oxide during VCD exposure resulted in a greater (P<0.05) loss of primordial and small primary follicles relative to VCD-treated ovaries. Also, relative to controls, meh mRNA was increased (P<0.05) on day 4 of VCD (30 µM) exposure, followed by increased (P<0.05) mEH protein after 6 days. Furthermore, inhibition of PI3K signaling increased mEH mRNA and protein expression. Thus, these results support a functional role for mEH in the rat ovary, and demonstrate the involvement of PI3K signaling in regulation of ovarian xenobiotic metabolism by mEH.


Subject(s)
Cyclohexenes/metabolism , Epoxide Hydrolases/physiology , Ovary/drug effects , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Vinyl Compounds/metabolism , Animals , Cyclohexenes/toxicity , Epoxide Hydrolases/genetics , Female , Ovary/enzymology , Phosphoinositide-3 Kinase Inhibitors , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Vinyl Compounds/toxicity
7.
Biol Reprod ; 85(4): 755-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21677306

ABSTRACT

In vitro exposure of Postnatal Day 4 (PND4) rat ovaries to the occupational chemical 4-vinylcyclohexene diepoxide (VCD) destroys specifically primordial and primary follicles via acceleration of atresia. Because oocyte-expressed c-kit (KIT) plays a critical role in follicle survival and activation, a direct interaction of VCD with KIT as its mechanism of ovotoxicity was investigated. PND4 rat ovaries were cultured with and without VCD (30 µM) for 2 days. When assessed by Western analysis or mobility shift detection, phosphorylated KIT (pKIT) was decreased (P < 0.05) by VCD exposure, while total KIT protein was unaffected. Anti-mouse KIT2 (ACK2) antibody binds KIT and blocks its signaling pathways, whereas anti-mouse KIT 4 (ACK4) antibody binds KIT but does not block its activity. PND4 rat ovaries were incubated for 2 days with and without VCD with and without ACK2 (80 µg/ml) or ACK4 (80 µg/ml). ACK2 decreased pKIT; however, ACK4 had no effect. Conversely, ACK2 did not affect a VCD-induced decrease in pKIT, whereas ACK4 further reduced it. Because ACK2 and ACK4 (known to directly bind KIT) affect VCD responses, these results support the fact that VCD interacts directly with KIT. The effect of these antibodies on VCD-induced follicle loss was measured after 8 days of incubation. ACK2 further reduced (P < 0.05) VCD-induced follicle loss, whereas ACK4 did not affect it. These findings demonstrate that VCD induces ovotoxicity by direct inhibition of KIT autophosphorylation of the oocyte. The data also further support the vital function of KIT and its signaling pathway in primordial follicle survival and activation, as well as its role in VCD-induced ovotoxicity.


Subject(s)
Cyclohexenes/toxicity , Environmental Pollutants/toxicity , Ovary/drug effects , Protein Kinase Inhibitors/toxicity , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Vinyl Compounds/toxicity , Animals , Animals, Newborn , Antibodies, Blocking/metabolism , Antigen-Antibody Reactions/drug effects , Cyclohexenes/antagonists & inhibitors , Environmental Pollutants/antagonists & inhibitors , Female , Follicular Atresia/drug effects , Ligands , Molecular Targeted Therapy , Molecular Weight , Organ Culture Techniques , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/growth & development , Ovary/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/agonists , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Inbred F344 , Vinyl Compounds/antagonists & inhibitors
8.
Biol Reprod ; 84(4): 743-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21076081

ABSTRACT

4-Vinylcyclohexene diepoxide (VCD), an occupational chemical that specifically destroys primordial and small primary follicles in the ovaries of rats and mice, is thought to target an oocyte-expressed tyrosine kinase receptor, Kit. This study compared the temporal effect of VCD on protein distribution of KIT and its downstream PIK3-activated proteins, AKT and FOXO3. Postnatal Day 4 Fischer 344 rat ovaries were cultured in control media ± VCD (30 µM) for 2-8 days (d2-d8). KIT, AKT, phosphorylated AKT, FOXO3, and pFOXO3 protein levels were assessed by Western blotting and/or immunofluorescence staining with confocal microscopy. Phosphorylated AKT was decreased (P < 0.05) in oocyte nuclei in primordial (39% decrease) and small primary (37% decrease) follicles within 2 days of VCD exposure. After d4, VCD reduced (P < 0.05) oocyte staining for KIT (primordial, 44% decrease; small primary, 39% decrease) and FOXO3 (primordial, 40% decrease; small primary, 36% decrease) protein. Total AKT and pFOXO3 were not affected by VCD at any time. Akt1 mRNA, as measured by quantitative RT-PCR, was reduced (P < 0.05) by 23% on d4 of VCD exposure, but returned to control levels on d6 and d8. VCD exposure reduced Foxo3a mRNA by 26% on d6 (P < 0.05) and by 23% on d8 (P < 0.1). These results demonstrate that the earliest observed effect of VCD is an inhibition of phosphorylation and nuclear localization of AKT in the oocyte of primordial and small primary follicles. This event is followed by reductions in KIT and FOXO3 protein subcellular distribution prior to changes in mRNA. Thus, these findings further support that VCD induces ovotoxicity by directly targeting the oocyte through posttranslational inhibition of KIT-mediated signaling components.


Subject(s)
Cyclohexenes/toxicity , Ovary/drug effects , Ovary/metabolism , Phosphoinositide-3 Kinase Inhibitors , Vinyl Compounds/toxicity , Animals , Base Sequence , DNA Primers/genetics , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Ovary/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Signal Transduction/drug effects
9.
Toxicol Appl Pharmacol ; 249(1): 1-7, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816688

ABSTRACT

Anti-Müllerian hormone (AMH) is produced by granulosa cells in primary to small antral follicles of the adult ovary and helps maintain primordial follicles in a dormant state. The industrial chemical, 4-vinylcyclohexene diepoxide (VCD) causes specific ovotoxicity in primordial and small primary follicles of mice and rats. Previous studies suggest that this ovotoxicity involves acceleration of primordial to primary follicle recruitment via interactions with the Kit/Kit ligand signaling pathway. Because of its accepted role in inhibiting primordial follicle recruitment, the present study was designed to investigate a possible interaction between AMH and VCD-induced ovotoxicity. Protein distribution of AMH was compared in neonatal and adult F344 rat ovaries. AMH protein was visualized by immunofluorescence microscopy in large primary and secondary follicles of the adult ovary, but in small primary follicles in neonatal rat ovaries. In cultured postnatal day (PND) 4 F344 rat ovaries, VCD exposure (30 µM, 2-8 days) decreased (P<0.05) AMH mRNA (d4-8) and protein (d6-8). Recombinant AMH (100-400 mg/ml) in PND4 ovaries cultured 8 days±VCD (30 µM) caused an increase (P<0.05) in primordial, and a decrease (P<0.05) in small primary follicles, supporting that AMH retarded primordial follicle recruitment. However, no concentration of AMH had an effect on VCD-induced ovotoxicity. Whereas, VCD caused a reduction in expression of AMH (d4-d8), it followed previously reported initial disruptions in Kit signaling induced by VCD (d2). Thus, collectively, these results do not support a mechanism whereby VCD causes ovotoxicity via generalized activation of primordial follicle recruitment, but instead provide further support for the specificity of other intracellular mechanisms involved in VCD-induced ovotoxicity.


Subject(s)
Anti-Mullerian Hormone/metabolism , Cyclohexenes/toxicity , Ovary/drug effects , Ovary/growth & development , Vinyl Compounds/toxicity , Animals , Animals, Newborn , Anti-Mullerian Hormone/analysis , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Female , Organ Culture Techniques , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/metabolism , Rats , Rats, Inbred F344 , Tissue Distribution
10.
Toxicol Appl Pharmacol ; 247(2): 71-5, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20542051

ABSTRACT

The occupational chemical 4-vinylcyclohexene diepoxide (VCD) selectively destroys ovarian small pre-antral follicles in rats and mice via apoptosis. Detoxification of VCD can occur through glutathione conjugation, catalyzed by glutathione S-transferase (GST) enzymes. Further, GST class pi (GSTp) can negatively regulate JNK activity through protein:protein interactions in extra-ovarian tissues. Dissociation of this protein complex in the face of chemical exposure releases the inhibition of pro-apoptotic JNK. Increased JNK activity during VCD-induced ovotoxicity has been shown in isolated ovarian small pre-antral follicles following in vivo dosing of rats (80mg/kg/day; 15days, i.p.). The present study investigated the pattern of ovarian GSTp expression during VCD exposure. Additionally, the effect of VCD on an ovarian GSTp:JNK protein complex was investigated. PND4 F344 rat ovaries were incubated in control medium+/-VCD (30muM) for 2-8days. VCD increased ovarian GSTp mRNA (P <0.05) relative to control on d4-d8; whereas GSTp protein was increased (P<0.05) on d6-d8. A GSTp:JNK protein complex was detected by immunoprecipitation and Western blotting in ovarian tissues. Relative to control, the amount of GSTp-bound JNK was increased (P=0.09), while unbound JNK was decreased (P<0.05) on d6 of VCD exposure. The VCD-induced decrease in unbound JNK was preceded by a decrease in phosphorylated c-Jun which occurred on d4. These findings are in support of a possible dual protective role for GSTp in the rat ovary, consisting of metabolism of VCD and inhibition of JNK-initiated apoptosis.


Subject(s)
Cyclohexenes/toxicity , Glutathione S-Transferase pi/metabolism , Ovary/enzymology , Vinyl Compounds/toxicity , Animals , Female , Glutathione S-Transferase pi/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Ovary/drug effects , Ovary/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344
11.
Comp Med ; 60(5): 380-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21262124

ABSTRACT

Chronic diseases including coronary heart disease and osteoporosis represent a substantial health burden to postmenopausal women, yet the initiation of these conditions and their relationships with reproductive aging remain poorly understood. This situation is due, in part, to the lack of animal models reflecting ovarian and hormonal characteristics of peri- and postmenopausal women. Ovaries of women approaching menopause are nearly depleted of primordial follicles but retain a pool of larger developing follicles and androgen-producing stroma, a condition known as reduced ovarian reserve (ROR). The long-term goal of the research presented here was to create a monkey model of reproductive aging, beginning with ROR and progressing to perimenopause and finally postmenopause. Here we sought to develop a method to reduce primordial follicles in cynomolgus monkeys (Macaca fascicularis) and document hormonal changes associated with follicle reduction or ROR. At 30 d after surgical placement of a biodegradable fiber containing approximately 200 mg of 4-vinlycyclohexene diepoxide (VCD) next to one ovary in each of 8 monkeys, primordial follicles were reduced by approximately 70%, with a corresponding decrease (83%) in antimüllerian hormone (AMH, a serum marker of ovarian follicle numbers). At 4 mo after VCD-treatment of both ovaries in 29 monkeys (approximately 200 mg VCD per ovary), AMH was reduced 56% from baseline, testosterone was unchanged, and follicular phase estradiol was slightly increased. These data indicate that VCD treatment markedly reduced primordial follicles while preserving larger estradiol- and testosterone-producing follicles and ovarian stroma, a condition that mimics ROR in women.


Subject(s)
Disease Models, Animal , Macaca fascicularis , Ovarian Diseases/chemically induced , Ovarian Follicle/drug effects , Sexual Development/drug effects , Animals , Anti-Mullerian Hormone/blood , Cyclohexenes , Female , Ovarian Follicle/pathology , Ovary/drug effects , Ovary/pathology , Vinyl Compounds
12.
Toxicol Appl Pharmacol ; 241(2): 127-34, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19695275

ABSTRACT

4-vinylcyclohexene diepoxide (VCD) is an ovotoxicant that specifically destroys primordial and small primary follicles in the ovaries of mice and rats. In contrast, 7,12-dimethylbenz[a]anthracene (DMBA) is ovotoxic to all ovarian follicle classes. This study investigated phosphatidylinositol-3 kinase signaling involvement in VCD- and DMBA-induced ovotoxicity. Postnatal day (PND) 4 Fischer 344 (F344) rat whole ovaries were cultured for 2-12 days in vehicle control, VCD (30 microM), or DMBA (1 microM), +/-PI3 kinase inhibitor LY294002 (20 microM) or its inactive analog LY303511 (20 microM). Following culture, ovaries were histologically evaluated, and healthy follicles were classified and counted. PI3 kinase inhibition had no effect on primordial follicle number, but reduced (P<0.05) small primary and larger follicles beginning on day 4. VCD caused primordial and small primary follicle loss (P<0.05) beginning on day 6. With PI3 kinase inhibition, VCD did not affect primordial follicles (P>0.05) at any time, but did cause loss (P<0.05) of small primary follicles. DMBA exposure caused primordial and small primary follicle loss (P<0.05) on day 6. Further, DMBA-induced primordial and small primary follicle loss was greater with PI3 kinase inhibition (P<0.05) than with DMBA alone. These results support that (1) PI3 kinase mediates primordial to small primary follicle recruitment, (2) VCD, but not DMBA, enhances ovotoxicity by increasing primordial to small primary follicle recruitment, and (3) in addition to xenobiotic-induced ovotoxicity, VCD is also a useful model chemical with which to elucidate signaling mechanisms involved in primordial follicle recruitment.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Chromones/pharmacology , Cyclohexenes/toxicity , Morpholines/pharmacology , Ovarian Follicle/drug effects , Phosphoinositide-3 Kinase Inhibitors , Vinyl Compounds/toxicity , Animals , Animals, Newborn , Female , In Vitro Techniques , Ovarian Diseases/chemically induced , Ovarian Diseases/pathology , Ovarian Follicle/pathology , Piperazines/pharmacology , Rats , Time Factors
13.
Biol Reprod ; 79(2): 318-27, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18448842

ABSTRACT

Repeated daily dosing of rats with the occupational chemical 4-vinylcyclohexene diepoxide (VCD) depletes the ovary of primordial and primary follicles through an increase in the natural process of atresia. Additionally, in vitro exposure of Postnatal Day 4 (PND 4) rat ovaries to VCD causes similar follicular depletion. This study was designed to investigate survival signaling pathways that may be associated with VCD-induced ovotoxicity in small preantral follicles. Female Fischer 344 rats (PND 28) were dosed daily (80 mg/kg/day VCD i.p.; 12 days in vivo), and PND 4 ovaries were cultured (VCD 20 or 30 microM; 8 days in vitro). Microarray analysis identified a subset of 14 genes whose expression was increased or decreased by VCD in both experiments (i.e., via both exposure routes). Particularly, the analysis showed that relative to controls, VCD did not affect mRNA expression of growth and differentiation factor 9 (Gdf9), whereas there were decreases in mRNA encoding bone morphogenic protein receptor 1a (Bmpr1a) and Kit. To confirm findings from microarray, the genes Gdf9, Bmpr1a, and Kit were further examined. When growth factors associated with these pathways were added to ovarian cultures during VCD exposure, GDF9 and BMP4 had no effect on VCD-induced ovotoxicity; however, KITL attenuated this follicle loss. Additionally, there was a decrease in Kit and an increase in Kitl expression (mRNA and protein) following VCD exposure, relative to control. These results support that VCD compromises KIT/KITL signaling, which is critical for follicular survival in primordial and primary follicles.


Subject(s)
Cyclohexenes/pharmacology , Follicular Atresia/drug effects , Follicular Atresia/genetics , Proto-Oncogene Proteins c-kit/physiology , Stem Cell Factor/physiology , Vinyl Compounds/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bone Morphogenetic Protein 15 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Female , Gene Expression Profiling , Growth Differentiation Factor 9 , Intercellular Signaling Peptides and Proteins/pharmacology , Oligonucleotide Array Sequence Analysis , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Proto-Oncogene Proteins c-kit/genetics , Rats , Rats, Inbred F344 , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cell Factor/genetics , Stem Cell Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...