Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Endocrinology ; 164(3)2023 01 09.
Article in English | MEDLINE | ID: mdl-36626144

ABSTRACT

Obesity, cardiometabolic disease, cognitive decline, and osteoporosis are symptoms of postmenopause, which can be modeled using 4-vinylcyclohexene diepoxide (VCD)-treated mice to induce ovarian failure and estrogen deficiency combined with high-fat diet (HFD) feeding. The trend of replacing saturated fatty acids (SFAs), for example coconut oil, with seed oils that are high in polyunsaturated fatty acids, specifically linoleic acid (LA), may induce inflammation and gut dysbiosis, and worsen symptoms of estrogen deficiency. To investigate this hypothesis, vehicle (Veh)- or VCD-treated C57BL/6J mice were fed a HFD (45% kcal fat) with a high LA:SFA ratio (22.5%: 8%), referred to as the 22.5% LA diet, or a HFD with a low LA:SFA ratio (1%: 31%), referred to as 1% LA diet, for a period of 23 to 25 weeks. Compared with VCD-treated mice fed the 22.5% LA diet, VCD-treated mice fed the 1% LA diet showed lower weight gain and improved glucose tolerance. However, VCD-treated mice fed the 1% LA diet had higher blood pressure and showed evidence of spatial cognitive impairment. Mice fed the 1% LA or 22.5% LA diets showed gut microbial taxa changes that have been associated with a mix of both beneficial and unfavorable cognitive and metabolic phenotypes. Overall, these data suggest that consuming different types of dietary fat from a variety of sources, without overemphasis on any particular type, is the optimal approach for promoting metabolic health regardless of estrogen status.


Subject(s)
Dietary Fats , Fatty Acids , Mice , Female , Animals , Coconut Oil , Mice, Inbred C57BL , Dietary Fats/adverse effects , Diet, High-Fat/adverse effects , Linoleic Acid , Homeostasis , Cognition , Estrogens
2.
Metabolism ; 83: 234-244, 2018 06.
Article in English | MEDLINE | ID: mdl-29522773

ABSTRACT

BACKGROUND: Menopause is associated with fatty liver, glucose dysregulation, increased body fat, and impaired bone quality. Previously, it was demonstrated that single sessions of high-intensity interval exercise (HIIE) are more effective than distance- and duration-matched continuous exercise (CE) on altering hepatic triglyceride (TG) metabolism and very-low density lipoprotein-TG (VLDL-TG) secretion. METHODS: Six weeks training using these modalities was examined for effects on hepatic TG metabolism/secretion, glucose tolerance, body composition, and bone mineral density (BMD) in ovariectomized (OVX) and sham-operated (SHAM) mice. OVX and SHAM were assigned to distance- and duration-matched CE and HIIE, or sedentary control. RESULTS: Energy expenditure during exercise was confirmed to be identical between CE and HIIE and both similarly reduced post-exercise absolute carbohydrate oxidation and spontaneous physical activity (SPA). OVX vs. SHAM displayed impaired glucose tolerance and greater body fat despite lower hepatic TG, and these outcomes were not affected by training. Only HIIE increased hepatic AMPK in OVX and SHAM, but neither training type impacted VLDL-TG secretion. As expected, BMD was lower in OVX, and training did not affect long bones. CONCLUSIONS: The results reveal intensity-dependent effects on hepatic AMPK expression and general exercise effects on subsequent SPA and substrate oxidation that is independent of estrogen status. These findings support the notion that HIIE can impact aspects of liver physiology in females while the effects of exercise on whole body substrate selection appear to be independent of training intensity. However, neither exercise approach mitigated the impairment in glucose tolerance and elevated body fat occurring in OVX mice.


Subject(s)
Energy Metabolism/physiology , Lipid Metabolism/physiology , Liver/metabolism , Motor Activity/physiology , Ovariectomy , Physical Conditioning, Animal/methods , Physical Conditioning, Animal/physiology , Animals , Estrogens/deficiency , Estrogens/pharmacology , Female , Mice , Mice, Inbred C57BL , Ovariectomy/adverse effects
3.
Reprod Toxicol ; 62: 18-26, 2016 07.
Article in English | MEDLINE | ID: mdl-27103539

ABSTRACT

Developmental exposure to endocrine-disrupting compounds (EDCs) alters reproduction and energy homeostasis, both of which are regulated by the arcuate nucleus (ARC). Little is known about the effects of EDC on ARC gene expression. In Experiment #1, pregnant dams were treated with either two doses of bisphenol A (BPA) or oil from embryonic day (E)18-21. Neonates were injected from postnatal day (PND)0-7. Vaginal opening, body weights, and ARC gene expression were measured. Chrm3 (muscarinic receptor 3) and Adipor1 (adiponectin receptor 1) were decreased by BPA. Bdnf (brain-derived neurotropic factor), Igf1 (insulin-like growth factor 1), Htr2c (5-hydroxytryptamine receptor), and Cck2r (cholescystokinin 2 receptor) were impacted. In Experiment #2, females were exposed to BPA, diethylstilbestrol (DES), di(2-ethylhexyl)phthalate, or methoxychlor (MXC) during E11-PND7. MXC and DES advanced the age of vaginal opening and ARC gene expression was impacted. These data indicate that EDCs alter ARC genes involved in reproduction and energy homeostasis in females.


Subject(s)
Endocrine Disruptors/toxicity , Gene Expression Regulation, Developmental/drug effects , Prenatal Exposure Delayed Effects , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Benzhydryl Compounds/toxicity , Brain-Derived Neurotrophic Factor/genetics , Diethylhexyl Phthalate/toxicity , Diethylstilbestrol/toxicity , Female , Homeostasis/drug effects , Insulin-Like Growth Factor I/genetics , Methoxychlor/toxicity , Phenols/toxicity , Pregnancy , Rats, Inbred F344 , Receptor, Cholecystokinin B/genetics , Receptor, Muscarinic M3/genetics , Receptor, Serotonin, 5-HT2C/genetics , Receptors, Adiponectin/genetics , Sexual Maturation/drug effects
4.
Reprod Toxicol ; 51: 72-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25549949

ABSTRACT

The major metabolite of the estrogenic pesticide methoxychlor (MXC) HPTE is a stronger ESR1 agonist than MXC and acts also as an ESR2 antagonist. In granulosa cells (GCs), FSH stimulates estradiol via the second messenger cAMP. HPTE inhibits estradiol biosynthesis, and this effect is greater in FSH-treated GCs than in cAMP-treated GCs. Therefore; we examined the effect of MXC/HPTE on FSH-stimulated cAMP production in cultured GCs. To test involvement of ESR-signaling, we used the ESR1 and ESR2 antagonist ICI 182,780, ESR2 selective antagonist PHTPP, and ESR2 selective agonist DPN. ESR1 and ESR2 mRNA and protein levels were quantified. Both HPTE and MXC inhibited the FSH-induced cAMP production. ICI 182,780 and PHTPP mimicked the inhibitory action of HPTE. MXC/HPTE reduced FSH-stimulated Esr2 mRNA and protein to basal levels. MXC/HPTE also inhibited FSH-stimulated Esr1. The greater inhibition on FSH-stimulated GCs is likely due to reduced cAMP level that involves ESR-signaling, through ESR2.


Subject(s)
Estrogen Receptor Modulators/pharmacology , Granulosa Cells/drug effects , Insecticides/pharmacology , Methoxychlor/pharmacology , Phenols/pharmacology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Fulvestrant , Granulosa Cells/metabolism , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats, Sprague-Dawley
5.
Calcif Tissue Int ; 95(6): 521-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25326143

ABSTRACT

Endogenous estrogen has beneficial effects on mature bone and negatively affects the developing skeleton, whereas the effect of environmental estrogens is not known. Methoxychlor (MXC) is a synthetic estrogen known as a persistent organochlorine and used as a pesticide. Methoxychlor and its metabolites display estrogenic, anti-estrogenic and anti-androgenic activity and may therefore influence bone. Fifty-eight male fetal and neonatal rats were exposed to either: a negative control (DMSO), 0.020, 100 mg/kg MXC, or 1 mg/kg ß-estradiol-3-benzoate (EB; positive control). Rats were treated daily for 11 days, from embryonic day 19 to postnatal day (PND) 7 or for 4 days during the postnatal period (PND 0-7). All rats were analyzed at PND-84. Total body, femur, spine, and tibia areal bone mineral density (BMD) and content (BMC), lean body mass (LBM) and fat were measured by dual energy X-ray absorptiometry. Bone geometry and volumetric (v) BMD were measured using micro-computed tomography and biomechanical properties using three-point bending were assessed. Rats exposed to EB or MXC (at either the high and/or low dose), independent of exposure interval showed lower body weight, LBM, tibia and femur BMD and length, and total body BMD and BMC than DMSO control group (p ≤ 0.05). Methoxychlor and EB exposure increased cortical porosity compared to DMSO controls. Trabecular vBMD, number and separation, and cortical polar moment of inertia and cross-sectional area were lower due to EB exposure compared to control (p < 0.05). Early MXC exposure compromises cortical porosity and bone size at maturity, and could ultimately increase the risk of fracture with aging.


Subject(s)
Bone Density/drug effects , Bone and Bones/drug effects , Insecticides/toxicity , Methoxychlor/toxicity , Prenatal Exposure Delayed Effects/pathology , Absorptiometry, Photon , Animals , Animals, Newborn , Body Composition/drug effects , Female , Fetus , Male , Pregnancy , Rats , Rats, Inbred F344
6.
Biol Reprod ; 88(2): 52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23303685

ABSTRACT

Transient exposure to methoxychlor (MXC), an environmental endocrine-disrupting chemical, during fetal and neonatal stages causes ovarian dysfunction in pubertal, adult, and aging animals. Adult animals have reduced number of ovulations and abnormal follicular composition associated with altered gene expression and DNA methylation patterns. To test the hypothesis that the ovarian epigenomic changes induced by MXC are detectable following the exposure period, leading to altered gene expression by adulthood, we conducted a targeted genome-wide methylation study using Nimblegen 3x720K CpG Island Plus RefSeq Promoter Arrays. Control (vehicle), low-dose MXC (20 µg/kg/day), or high-dose MXC (100 mg/kg/day) treatments were administered between Embryonic Day 19 and Postnatal Day (PND) 7. Ovaries were collected at PND 7 immediately after exposure or at adulthood, PND 60. Array hybridizations were conducted with genomic DNA after methylated DNA immunoprecipitation and the array data were analyzed. DNA methylation events were functionally annotated, and candidate loci common to all the treatments or unique to some treatments were identified. Specific loci encoding signaling molecules such as the regulatory subunit p85 of phosphoinositide-3-kinase, insulin-like growth factor-1 receptor, Harvey rat sarcoma viral oncogene, insulin receptor, and forkhead box protein O3 were identified to be hypermethylated in MXC-treated ovaries at PND 7 and/or PND 60. Examination of gene expression changes with TaqMan low-density arrays revealed that nearly 25% of the genes that were assayed were downregulated. These data demonstrate that key molecules in specific signaling pathways such as PTEN signaling, IGF-1 signaling, or rapid estrogen signaling are epigenetically altered in MXC-exposed ovaries, which is associated with ovarian dysfunction and female infertility.


Subject(s)
DNA Methylation/drug effects , Endocrine Disruptors/pharmacology , Genome/genetics , Ovary/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Signal Transduction/physiology , Transcriptome/drug effects , Animals , DNA Methylation/genetics , Dose-Response Relationship, Drug , Female , Genome/drug effects , Infertility, Female , Methoxychlor/pharmacology , Models, Animal , Ovarian Follicle/drug effects , Ovarian Follicle/physiopathology , Ovary/drug effects , Ovary/embryology , Ovulation/drug effects , Ovulation/physiology , Pregnancy , Rats , Rats, Inbred F344 , Signal Transduction/drug effects , Transcriptome/genetics
7.
Mol Endocrinol ; 25(12): 2157-68, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22016562

ABSTRACT

Gestational exposure to the estrogenic endocrine disruptor methoxychlor (MXC) disrupts the female reproductive system at the molecular, physiological, and behavioral levels in adulthood. The current study addressed whether perinatal exposure to endocrine disruptors re-programs expression of a suite of genes expressed in the hypothalamus that control reproductive function and related these molecular changes to premature reproductive aging. Fischer rats were exposed daily for 12 consecutive days to vehicle (dimethylsulfoxide), estradiol benzoate (EB) (1 mg/kg), and MXC (low dose, 20 µg/kg or high dose, 100 mg/kg), beginning on embryonic d 19 through postnatal d 7. The perinatally exposed females were aged to 16-17 months and monitored for reproductive senescence. After euthanasia, hypothalamic regions [preoptic area (POA) and medial basal hypothalamus] were dissected for real-time PCR of gene expression or pyrosequencing to assess DNA methylation of the Esr1 gene. Using a 48-gene PCR platform, two genes (Kiss1 and Esr1) were significantly different in the POA of endocrine-disrupting chemical-exposed rats compared with vehicle-exposed rats after Bonferroni correction. Fifteen POA genes were up-regulated by at least 50% in EB or high-dose MXC compared with vehicle. To understand the epigenetic basis of the increased Esr1 gene expression, we performed bisulfite conversion and pyrosequencing of the Esr1 promoter. EB-treated rats had significantly higher percentage of methylation at three CpG sites in the Esr1 promoter compared with control rats. Together with these molecular effects, perinatal MXC and EB altered estrous cyclicity and advanced reproductive senescence. Thus, early life exposure to endocrine disruptors has lifelong effects on neuroendocrine gene expression and DNA methylation, together with causing the advancement of reproductive senescence.


Subject(s)
Endocrine Disruptors/pharmacology , Estradiol/analogs & derivatives , Menopause, Premature/drug effects , Methoxychlor/pharmacology , Preoptic Area/metabolism , Animals , Animals, Newborn , Base Sequence , Body Weight/drug effects , CpG Islands , DNA Methylation , Estradiol/blood , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrous Cycle/drug effects , Estrous Cycle/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Kisspeptins/genetics , Kisspeptins/metabolism , Maternal-Fetal Exchange , Menopause, Premature/genetics , Molecular Sequence Data , Pregnancy , Preoptic Area/drug effects , Progesterone/blood , Promoter Regions, Genetic , Rats , Rats, Inbred F344 , Regulatory Sequences, Nucleic Acid , Up-Regulation
8.
Front Neuroendocrinol ; 31(4): 420-39, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20609371

ABSTRACT

The link between in utero and neonatal exposure to environmental toxicants, such as endocrine-disrupting chemicals (EDCs) and adult female reproductive disorders is well established in both epidemiological and animal studies. Recent studies examining the epigenetic mechanisms involved in mediating the effects of EDCs on female reproduction are gathering momentum. In this review, we describe the developmental processes that are susceptible to EDC exposures in female reproductive system, with a special emphasis on the ovary. We discuss studies with select EDCs that have been shown to have physiological and correlated epigenetic effects in the ovary, neuroendocrine system, and uterus. Importantly, EDCs that can directly target the ovary can alter epigenetic mechanisms in the oocyte, leading to transgenerational epigenetic effects. The potential mechanisms involved in such effects are also discussed.


Subject(s)
Endocrine Disruptors/toxicity , Epigenomics , Infertility, Female/chemically induced , Ovary/drug effects , Animals , Environmental Pollutants/toxicity , Female , Gene Expression Profiling , Genitalia, Female/drug effects , Genitalia, Female/growth & development , Humans , Infertility, Female/genetics , Male , Mice , Neurosecretory Systems/drug effects , Neurosecretory Systems/growth & development , Ovary/growth & development , Receptors, Androgen/analysis , Receptors, Estrogen/analysis , Uterus/drug effects , Uterus/growth & development
9.
Endocrinology ; 150(10): 4681-91, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19589859

ABSTRACT

Exposure to endocrine-disrupting chemicals during development could alter the epigenetic programming of the genome and result in adult-onset disease. Methoxychlor (MXC) and its metabolites possess estrogenic, antiestrogenic, and antiandrogenic activities. Previous studies showed that fetal/neonatal exposure to MXC caused adult ovarian dysfunction due to altered expression of key ovarian genes including estrogen receptor (ER)-beta, which was down-regulated, whereas ERalpha was unaffected. The objective of the current study was to evaluate changes in global and gene-specific methylation patterns in adult ovaries associated with the observed defects. Rats were exposed to MXC (20 microg/kgxd or 100 mg/kg.d) between embryonic d 19 and postnatal d 7. We performed DNA methylation analysis of the known promoters of ERalpha and ERbeta genes in postnatal d 50-60 ovaries using bisulfite sequencing and methylation-specific PCRs. Developmental exposure to MXC led to significant hypermethylation in the ERbeta promoter regions (P < 0.05), whereas the ERalpha promoter was unaffected. We assessed global DNA methylation changes using methylation-sensitive arbitrarily primed PCR and identified 10 genes that were hypermethylated in ovaries from exposed rats. To determine whether the MXC-induced methylation changes were associated with increased DNA methyltransferase (DNMT) levels, we measured the expression levels of Dnmt3a, Dnmt3b, and Dnmt3l using semiquantitative RT-PCR. Whereas Dnmt3a and Dnmt3l were unchanged, Dnmt3b expression was stimulated in ovaries of the 100 mg/kg MXC group (P < 0.05), suggesting that increased DNMT3B may cause DNA hypermethylation in the ovary. Overall, these data suggest that transient exposure to MXC during fetal and neonatal development affects adult ovarian function via altered methylation patterns.


Subject(s)
DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Methoxychlor/toxicity , Ovary/drug effects , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn , DNA Modification Methylases/metabolism , Down-Regulation , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Ovary/metabolism , Pregnancy , Promoter Regions, Genetic , Rats , Rats, Inbred F344 , Up-Regulation
10.
Toxicol Sci ; 110(1): 95-106, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19414516

ABSTRACT

Ovarian granulosa cells play a central role in steroidogenesis, which is critical for female reproduction. Follicle-stimulating hormone (FSH) promotes cyclic adenosine monophosphate (cAMP)-mediated signaling to regulate granulosa cell steroidogenesis. We have shown previously that 2,2-bis-(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE) inhibits FSH- and dibutyryl cAMP-stimulated steroidogenesis and affects the messenger RNA levels of steroidogenic pathway enzymes in rat granulosa cells. However, HPTE showed a differential effect in FSH- and cAMP-stimulated cells in that HPTE more completely blocked FSH- when compared to cAMP-driven steroidogenesis. The objective of this study was to analyze the effects of HPTE on global gene expression profiles in untreated granulosa cells and those challenged with FSH or cAMP. Granulosa cells from immature rats were cultured with 0, 1, 5, or 10 microM HPTE in the presence or absence of either 3 ng FSH/ml or 1mM cAMP for 48 h. Total RNA was isolated for real-time quantitative PCR and microarray analysis using the GeneChip Rat Genome 230 2.0 and ArrayAssist Microarray Suite. An investigation of changes in gene expression across all HPTE treatments showed that HPTE altered more genes in FSH- (approximately 670 genes) than in cAMP-stimulated cells (approximately 366 genes). Analysis confirmed that HPTE more effectively inhibited FSH- than cAMP-induced steroid pathway gene expression and steroidogenesis. Furthermore, expression patterns of novel genes regulating signal transduction, transport, cell cycle, adhesion, differentiation, motility and growth, apoptosis, development, and metabolism were all altered by HPTE. This study further established that HPTE exerts differential effects within the granulosa cell steroidogenic pathway and revealed that these effects include broader changes in gene expression.


Subject(s)
Gene Expression Profiling , Granulosa Cells/drug effects , Insecticides/metabolism , Methoxychlor/metabolism , Ovary/cytology , Phenols/toxicity , Animals , Cells, Cultured , Cyclic AMP/pharmacology , Estradiol/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Models, Genetic , Oligonucleotide Array Sequence Analysis , Ovary/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
11.
Toxicol Appl Pharmacol ; 233(2): 286-96, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18848953

ABSTRACT

Methoxychlor (MXC) is an organochlorine pesticide with estrogenic, anti-estrogenic, and anti-androgenic properties. To investigate whether transient developmental exposure to MXC could cause adult ovarian dysfunction, we exposed Fischer rats to 20 microg/kg/day (low dose; environmentally relevant dose) or 100 mg/kg/day (high dose) MXC between 19 days post coitum and postnatal day 7. Multiple reproductive parameters, serum hormone levels, and ovarian morphology and molecular markers were examined from prepubertal through adult stages. High dose MXC accelerated pubertal onset and first estrus, reduced litter size, and increased irregular cyclicity (P<0.05). MXC reduced superovulatory response to exogenous gonadotropins in prepubertal females (P<0.05). Rats exposed to high dose MXC had increasing irregular estrous cyclicity beginning at 4 months of age, with all animals showing abnormal cycles by 6 months. High dose MXC reduced serum progesterone, but increased luteinizing hormone (LH). Follicular composition analysis revealed an increase in the percentage of preantral and early antral follicles and a reduction in the percentage of corpora lutea in high dose MXC-treated ovaries (P<0.05). Immunohistochemical staining and quantification of the staining intensity showed that estrogen receptor beta was reduced by high dose MXC while anti-Mullerian hormone was upregulated by both low- and high dose MXC in preantral and early antral follicles (P<0.05). High dose MXC significantly reduced LH receptor expression in large antral follicles (P<0.01), and down-regulated cytochrome P450 side-chain cleavage. These results demonstrated that developmental MXC exposure results in reduced ovulation and fertility and premature aging, possibly by altering ovarian gene expression and folliculogenesis.


Subject(s)
Gene Expression Regulation/drug effects , Insecticides/toxicity , Methoxychlor/toxicity , Ovarian Follicle/drug effects , Ovulation/drug effects , Animals , Anti-Mullerian Hormone/metabolism , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Dose-Response Relationship, Drug , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/metabolism , Estrous Cycle/drug effects , Female , Fertility/drug effects , Immunohistochemistry , Insecticides/administration & dosage , Litter Size/drug effects , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Methoxychlor/administration & dosage , Ovarian Follicle/pathology , Pregnancy , Progesterone/metabolism , Rats , Rats, Inbred F344 , Sexual Maturation/drug effects
12.
Reprod Toxicol ; 26(3-4): 191-6, 2008.
Article in English | MEDLINE | ID: mdl-18848623

ABSTRACT

The rat is one of the most commonly used experimental animal species in biomedical research. The availability of new research tools in rats could therefore provide considerable advances in the areas where this mammal is extensively used. We report the development of a new green fluorescent protein (GFP) rat strain suitable for organ transplantation and the birth of GFP rats following orthotopic transplantation of neonatal ovaries from this newly developed GFP rat strain to a wild-type Fischer 344 (F344) strain. A new GFP rat strain was developed by backcrossing eGFP Sprague-Dawley (SD-Tg(CAG-EGFP)Cz-004Osb) to wild-type F344 for eight generations. Whole ovaries from postnatal day (PND) 8 or PND 21 GFP rats were transplanted orthotopically to bilaterally ovariectomized wild-type adult females (n=6). All recipients were mated, and three of the five resulting litters contained GFP pups. In the PND 8 group, all recipients cycled regularly and the ovarian morphology appeared normal when collected at 9 months post-transplantation. In the PND 21 group, 60% of the recipients displayed regular estrous cycles at 9 months post-transplantation, but showed reduced ovarian size. This new strain and neonatal orthotopic transplantation could be useful for many biomedical fields including transplantation, development, and reproductive toxicology.


Subject(s)
Green Fluorescent Proteins , Ovary/transplantation , Animals , Animals, Newborn , Female , Fertility , Ovary/drug effects , Ovary/growth & development , Rats , Rats, Inbred F344
13.
Mol Cell Endocrinol ; 294(1-2): 70-80, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18790002

ABSTRACT

The role transforming growth factor beta (TGFb) isoforms TGFb1, TGFb2 and TGFb3 have in the regulation of embryonic gonadal development was investigated with the use of null-mutant (i.e. knockout) mice for each of the TGFb isoforms. Late embryonic gonadal development was investigated because homozygote TGFb null-mutant mice generally die around birth, with some embryonic loss as well. In the testis, the TGFb1 null-mutant mice had a decrease in the number of germ cells at birth, postnatal day 0 (P0). In the testis, the TGFb2 null-mutant mice had a decrease in the number of seminiferous cords at embryonic day 15 (E15). In the ovary, the TGFb2 null-mutant mice had an increase in the number of germ cells at P0. TGFb isoforms appear to have a role in gonadal development, but interactions between the isoforms is speculated to compensate in the different TGFb isoform null-mutant mice.


Subject(s)
Embryonic Development/genetics , Mutation/genetics , Ovary/embryology , Ovary/metabolism , Testis/embryology , Testis/metabolism , Transforming Growth Factor beta/deficiency , Animals , Antigens, Nuclear/metabolism , Apoptosis , Cell Count , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Genotype , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Oligonucleotide Array Sequence Analysis , Ovary/cytology , Phenotype , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/embryology , Seminiferous Tubules/metabolism , Sex Ratio , Testis/cytology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/deficiency , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/deficiency , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
14.
J Endocrinol ; 195(3): 359-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18000299

ABSTRACT

The hepatocyte growth factor (HGF) system comprises HGF, its receptor (the c-met tyrosine kinase), HGF activator (HGFA) protein, and HGFA inhibitor (HAI). The components of the HGF system have been identified in a plethora of tissues to include the ovary and testis. In its traditional context, the HGF system works via paracrine- and autocrine-mediated feedback in which HGF (of mesenchymal origin) binds and activates c-met (within epithelial cells); target cells then respond to HGF via any number of morphogenic and functional changes. The concomitant presence of HGFA and HAI suggests that HGF bioactivity can be locally modulated. A number of studies have collectively shown that the mammalian ovary and testis contain HGF, c-met, and HGFA; very little is currently known regarding HAI within the gonad. Within the ovary, HGF controls numerous key functions which collectively regulate the growth and differentiation of ovarian follicles; these include cell growth, steroidogenesis, and apoptosis within theca cells and/or granulosa cells. Comparatively, less is known about the function of HGF within the testicular Leydig and Sertoli cells, but evidence is emerging that HGF may regulate somatic cell function, including Leydig cell steroidogenesis. Changes in the cellular origin of HGF and c-met during fetal and postnatal testicular development suggest that HGF, in collaboration with other growth factors, may regulate important aspects of testicular cell morphogenesis and differentiation which enable male sexual viability. Likewise, experimental evidence showing that HGF can modulate many vital processes which enable ovarian follicle growth, differentiation, and function indicate the importance of HGF in female reproduction. This review presents what is currently known regarding the expression of the HGF system and its function within the ovary and testis.


Subject(s)
Hepatocyte Growth Factor/physiology , Ovary/physiology , Testis/physiology , Animals , Apoptosis/physiology , Cell Division/physiology , Female , Gonadal Steroid Hormones/biosynthesis , Granulosa Cells/cytology , Granulosa Cells/physiology , Humans , Leydig Cells/metabolism , Male , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovary/cytology , Ovary/metabolism , Theca Cells/cytology , Theca Cells/physiology
15.
Reproduction ; 134(3): 455-72, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17709564

ABSTRACT

Gene expression profiles during sex determination and gonadal differentiation were investigated to identify new potential regulatory factors. Embryonic day 13 (E13), E14, and E16 rat testes and ovaries were used for microarray analysis, as well as E13 testis organ cultures that undergo testis morphogenesis and develop seminiferous cords in vitro. A list of 109 genes resulted from a selective analysis for genes present in male gonadal development and with a 1.5-fold change in expression between E13 and E16. Characterization of these 109 genes potentially important for testis development revealed that cytoskeletal-associated proteins, extracellular matrix factors, and signaling factors were highly represented. Throughout the developmental period (E13-E16), sex-enriched transcripts were more prevalent in the male with 34 of the 109 genes having testis-enriched expression during sex determination. In ovaries, the total number of transcripts with a 1.5-fold change in expression between E13 and E16 was similar to the testis, but none of those genes were both ovary enriched and regulated during the developmental period. Genes conserved in sex determination were identified by comparing changing transcripts in the rat analysis herein, to transcripts altered in previously published mouse studies of gonadal sex determination. A comparison of changing mouse and rat transcripts identified 43 genes with species conservation in sex determination and testis development. Profiles of gene expression during E13-E16 rat testis and ovary development are presented and candidate genes for involvement in sex determination and testis differentiation are identified. Analysis of cellular pathways did not reveal any specific pathways involving multiple candidate genes. However, the genes and gene network identified influence numerous cellular processes with cellular differentiation, proliferation, focal contact, RNA localization, and development being predominant.


Subject(s)
Gene Expression Regulation, Developmental , Sex Determination Processes , Testis/embryology , Animals , Computational Biology , Embryonic Development/genetics , Female , Gene Expression Profiling , Gene Regulatory Networks , Male , Morphogenesis/genetics , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Ovary/embryology , Rats , Rats, Sprague-Dawley , Sex Differentiation , Transcription, Genetic/physiology
16.
Reprod Toxicol ; 23(3): 337-52, 2007.
Article in English | MEDLINE | ID: mdl-17140764

ABSTRACT

Female reproductive function depends upon the exquisite control of ovarian steroidogenesis that enables folliculogenesis, ovulation, and pregnancy. These mechanisms are set during fetal and/or neonatal development and undergo phases of differentiation throughout pre- and post-pubescent life. Ovarian development and function are collectively regulated by a host of endogenous growth factors, cytokines, gonadotropins, and steroid hormones as well as exogenous factors such as nutrients and environmental agents. Endocrine disruptors represent one class of environmental agent that can impact female fertility by altering ovarian development and function, purportedly through estrogenic, anti-estrogenic, and/or anti-androgenic effects. This review discusses ovarian development and function and how these processes are affected by some of the known estrogenic and anti-androgenic endocrine disruptors. Recent information suggests not only that exposure to endocrine disruptors during the developmental period causes reproductive abnormalities in adult life but also that these abnormalities are transgenerational. This latter finding adds another level of importance for identifying and understanding the mechanisms of action of these agents.


Subject(s)
Endocrine Disruptors/poisoning , Environmental Pollutants/poisoning , Ovary/drug effects , Reproduction/drug effects , Animals , Embryonic Development/drug effects , Endocrine Disruptors/chemistry , Environmental Pollutants/chemistry , Female , Humans , Ovary/embryology , Pregnancy , Prenatal Exposure Delayed Effects , Reproduction/physiology
17.
J Endocrinol ; 191(3): 549-58, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17170213

ABSTRACT

Methoxychlor [1,1,1-trichloro-2,2-bis(4-methoxyphenyl) ethane; MXC] is a chlorinated hydrocarbon pesticide commonly used in the United States as a replacement for DDT [1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane]. While MXC is a weak estrogenic compound, its more active, major metabolite [2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane; HPTE] shows estrogenic, anti-estrogenic, or anti-androgenic properties depending on the receptor subtype with which it interacts. Anti-Mullerian hormone (AMH) is a paracrine factor that suppresses initial follicle recruitment in the ovary. Studies have shown the effects of exposure to MXC on adult ovarian morphology and function. However, the effect of exposure to MXC at an early postnatal stage on pre-pubertal follicular development and ovarian AMH production has not been studied. Around postnatal day (P) 4, most of the primordial follicular assembly in rats is complete, and a large number of primordial follicles transition into the primary follicle stage, a process that is inhibited by estrogen. The objective of this study was to examine the effect of early postnatal (P3-P10) MXC exposure on ovarian morphology and size, follicle number, and AMH production in the pre-pubertal (P20) rat ovary and to investigate the effect of HPTE on AMH production in immature rat granulosa cells in vitro. Female rats were injected (s.c.) daily with vehicle (control) or 1, 10, 50, 100, or 500 mg MXC/kg per day (referred to here as 1MXC, 10MXC, and so forth.) between P3 and P10. On P20, uterine and ovarian weights were determined, ovarian histology was examined, and follicles were counted and classified into primordial, primary, secondary, pre-antral, or antral stages using the two largest serial sections at the center of the ovary. Ovarian AMH production was examined using immunohistochemistry and western blot analysis. The effect of HPTE (0.5-25 microM) on AMH production in cultured immature rat granulosa cells was determined by western blot analysis. Ovarian weight was reduced by 50, 100, and 500MXC (P < 0.01). MXC treatment inhibited folliculogenesis. Both 100 and 500MXC had a reduced number of antral follicles (P < 0.05) with a concomitant increase in pre-antral follicles (P < 0.05). Follicle numbers were not significantly affected by 1, 10, or 50MXC. Total follicle number and the number of primordial, primary, or secondary stage follicles were not significantly different in all treatment groups. Immunohistochemistry showed that MXC-treated ovaries had more AMH-positive follicles with stronger AMH immunostaining. Western blot analysis showed that AMH production was 1.6 +/- 0.2, 1.85 +/- 0.6, and 2.2 +/- 0.5 times higher in the 50, 100, and 500MXC ovaries as compared with the control ovaries respectively (P < 0.05). Granulosa cells treated with 1 or 5 microM HPTE had significantly greater AMH production (P < 0.05). These results demonstrate that MXC inhibits early ovarian development and stimulates AMH production directly in the rat ovary. In addition, HPTE was shown to stimulate AMH production in rat granulosa cells. Endocrine disruptors are widespread in the environment, and MXC represents a model endocrine disruptor due to the multiple actions of its metabolites. This study confirms that the endocrine disruptor MXC inhibits follicular development and demonstrates for the first time that MXC and HPTE directly stimulate AMH production in the ovary. This novel finding suggests that elevated AMH may play a role in MXC's inhibitory effect in the ovary.


Subject(s)
Glycoproteins/biosynthesis , Insecticides/toxicity , Methoxychlor/toxicity , Ovary/metabolism , Testicular Hormones/biosynthesis , Animals , Animals, Newborn , Anti-Mullerian Hormone , Blotting, Western/methods , Female , Glycoproteins/analysis , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Immunohistochemistry/methods , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Ovarian Follicle/physiology , Ovary/drug effects , Ovary/pathology , Phenols/toxicity , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Testicular Hormones/analysis
18.
Reproduction ; 132(2): 291-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16885537

ABSTRACT

Hepatocyte growth factor (HGF) regulates granulosa cell (GC) steroidogenesis and suppresses apoptosis in non-ovarian cells. The hypothesis was thus developed that intraovarian HGF supports folliculogenesis by mediating steroidogenesis and suppressing apoptosis. To investigate the latter, the anti-apoptotic actions of HGF were tested in GCs and follicles isolated from immature rats. Results showed that HGF suppressed apoptosis in GC and follicle cultures as visualized using apoptosis indicator dye, YO-PRO-1. Immunohistochemistry was used to investigate the distribution of HGF, c-met, and HGF activator (HGFA) protein during folliculogenesis in equine chorionic gonadotropin (eCG)-primed rats. Immunoreactive HGF content was the greatest in GCs within preantral follicles. Following eCG, large antral follicles showed elevated HGF staining in theca and interstitial cells when compared with GCs. Intense c-met staining was observed in GCs within non-primed small preantral follicles; following eCG, the level of c-met was diminished in GCs, but increased within theca and interstitial cells. Theca, interstitium, and GCs in non-primed and primed ovaries contained HGFA. Following eCG, HGFA was more apparent in theca cells and the interstitium when compared to that in GCs within large antral follicles. The presence of HGF, c-met, and HGFA in preantral follicles would potentially enable the anti-apoptotic effects of HGF that were observed in vitro to occur in vivo. Advanced folliculogenesis led to a change in the cellular distribution of the HGF, c-met, and HGFA, suggesting that the ovarian HGF system is hormonally regulated in vivo.


Subject(s)
Hepatocyte Growth Factor/analysis , Ovarian Follicle/physiology , Ovary/chemistry , Animals , Apoptosis/drug effects , Blotting, Western/methods , Female , Granulosa Cells/cytology , Granulosa Cells/drug effects , Hepatocyte Growth Factor/pharmacology , Immunohistochemistry/methods , Ovarian Follicle/chemistry , Proto-Oncogene Proteins c-met/analysis , Rats , Rats, Sprague-Dawley , Serine Endopeptidases/analysis
19.
J Androl ; 27(6): 868-79, 2006.
Article in English | MEDLINE | ID: mdl-16837734

ABSTRACT

The current study was designed to examine the actions of a model endocrine disruptor on embryonic testis development and male fertility. Pregnant rats (F0) that received a transient embryonic exposure to an environmental endocrine disruptor, vinclozolin, had male offspring (F1) with reduced spermatogenic capacity. The reduced spermatogenetic capacity observed in the F1 male offspring was transmitted to the subsequent generations (F2-F4). The administration of vinclozolin, an androgen receptor antagonist, at 100 mg/kg/day from embryonic day 8-14 (E8-E14) of pregnancy to only the F0 dam resulted in a transgenerational phenotype in the subsequent male offspring in the F1-F4 generations. The litter size and male/female sex ratios were similar in controls and the vinclozolin generations. The average testes/body weight index of the postnatal day 60 (P60) males was not significantly different in the vinclozolin-treated generations compared to the controls. However, the testicular spermatid number, as well as the epididymal sperm number and motility, were significantly reduced in the vinclozolin generations compared to the control animals. Postnatal day 20 (P20) testis from the vinclozolin F2 generation had no morphological abnormalities, but did have an increase in spermatogenic cell apoptosis. Although the P60 testis morphology was predominantly normal, the germ cell apoptosis was significantly increased in the testes cross sections of animals from the vinclozolin generations. The increase in apoptosis was stage-specific in the testis, with tubules at stages IX-XIV having the highest increase in apoptotic germ cells. The tubules at stages I-V also had an increase in apoptotic germ cells compared to the control samples, but tubules at stages VI-VIII had no increase in apoptotic germ cells. An outcross of a vinclozolin generation male with a wild-type female demonstrated that the reduced spermatogenic cell phenotype was transmitted through the male germ line. An outcross with a vinclozolin generation female with a wild-type male had no phenotype. A similar phenotype was observed in outbred Sprague Dawley and inbred Fisher rat strains. Observations demonstrate that a transient exposure at the time of male sex determination to the antiandrogenic endocrine disruptor vinclozolin can induce an apparent epigenetic transgenerational phenotype with reduced spermatogenic capacity.


Subject(s)
Endocrine Disruptors/pharmacology , Oxazoles/pharmacology , Prenatal Exposure Delayed Effects , Spermatogenesis/drug effects , Animals , Apoptosis/drug effects , Epigenesis, Genetic/drug effects , Female , Litter Size/drug effects , Male , Pregnancy , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Testis/physiology
20.
Reprod Toxicol ; 22(4): 659-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16737795

ABSTRACT

The exquisitely balanced hormonal mechanisms that control female fertility can be affected by several internal and external factors including pathogens, genetic maladies, and environmental agents. In the latter group are natural and synthetic agents known as endocrine disruptors. One such compound, 2,2-bis-(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE), is the predominant metabolite of the pesticide methoxychlor. The effects of HPTE on ovarian steroidogenesis have not been previously reported and were investigated in the present study. Granulosa cells harvested from immature rats were treated with follicle-stimulating hormone (FSH) or N(6),2'-O-dibutyryladenosine 3',5'-cyclic monophosphate (db-cAMP) in the presence or absence of HPTE. After 48h, progesterone (P4) and estradiol-17beta (E2) concentrations were measured in the culture media. Steady-state levels of the mRNAs encoding steroidogenic acute regulatory protein (StAR), P450 side-chain cleavage (P450scc), 3beta-hydroxysteroid dehydrogenase type 1 (3beta-HSD), and P450 aromatase (P450arom) were examined using real-time PCR. Both FSH- and db-cAMP-stimulated P(4) accumulation were impaired by HPTE. In contrast, FSH-, but not db-cAMP-stimulated, E2 content was suppressed by HPTE. The FSH-dependent increase in the abundance of P450scc, 3beta-HSD, and P450arom mRNAs was blocked by HPTE; however, StAR expression was not altered. Although db-cAMP-dependent P450arom was moderately reduced by HPTE, the levels of db-cAMP-dependent StAR, P450scc, and 3beta-HSD mRNAs were increased in the presence of HPTE. These data collectively show that HPTE can disrupt P4 and E2 production in granulosa cells, with implications for sites of action both preceding and following the generation of cAMP. The steroid-modulatory effects of HPTE in granulosa cells appear to involve the general suppression of the FSH-dependent expression of mRNAs encoding steroid pathway proteins, whereas the disparate effects of HPTE on cAMP-dependent mRNA content in this regard suggest a broader and more complex mechanism of action.


Subject(s)
Granulosa Cells/drug effects , Methoxychlor/metabolism , Phenols/toxicity , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Aromatase/genetics , Cells, Cultured , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Estradiol/biosynthesis , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Granulosa Cells/metabolism , Phenols/metabolism , Phosphoproteins/genetics , Progesterone/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...