Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39166489

ABSTRACT

Chromatin changes in response to estrogen and progesterone are well established in cultured cells, but how they control gene expression under physiological conditions is largely unknown. To address this question, we examined in vivo estrous cycle dynamics of mouse uterus hormone receptor occupancy, chromatin accessibility and chromatin structure by combining RNA-seq, ATAC-seq, HiC-seq and ChIP-seq. Two estrous cycle stages were chosen for these analyses, diestrus (highest estrogen) and estrus (highest progesterone). Unexpectedly, rather than alternating with each other, estrogen receptor alpha (ERα) and progesterone receptor (PGR) were co-bound during diestrus and lost during estrus. Motif analysis of open chromatin followed by hypoxia inducible factor 2A (HIF2A) ChIP-seq and conditional uterine deletion of this transcription factor revealed a novel role for HIF2A in regulating diestrus gene expression patterns that were independent of either ERα or PGR binding. Proteins in complex with ERα included PGR and cohesin, only during diestrus. Combined with HiC-seq analyses, we demonstrate that complex chromatin architecture changes including enhancer switching are coordinated with ERα and PGR co-binding during diestrus and non-hormone receptor transcription factors such as HIF2A during estrus to regulate most differential gene expression across the estrous cycle.

2.
PLoS Biol ; 21(10): e3002334, 2023 10.
Article in English | MEDLINE | ID: mdl-37856394

ABSTRACT

Tissue development entails genetically programmed differentiation of immature cell types to mature, fully differentiated cells. Exposure during development to non-mutagenic environmental factors can contribute to cancer risk, but the underlying mechanisms are not understood. We used a mouse model of endometrial adenocarcinoma that results from brief developmental exposure to an estrogenic chemical, diethylstilbestrol (DES), to determine causative factors. Single-cell RNA sequencing (scRNAseq) and spatial transcriptomics of adult control uteri revealed novel markers of uterine epithelial stem cells (EpSCs), identified distinct luminal and glandular progenitor cell (PC) populations, and defined glandular and luminal epithelium (LE) cell differentiation trajectories. Neonatal DES exposure disrupted uterine epithelial cell differentiation, resulting in a failure to generate an EpSC population or distinguishable glandular and luminal progenitors or mature cells. Instead, the DES-exposed epithelial cells were characterized by a single proliferating PC population and widespread activation of Wnt/ß-catenin signaling. The underlying endometrial stromal cells had dramatic increases in inflammatory signaling pathways and oxidative stress. Together, these changes activated phosphoinositide 3-kinase/AKT serine-threonine kinase signaling and malignant transformation of cells that were marked by phospho-AKT and the cancer-associated protein olfactomedin 4. Here, we defined a mechanistic pathway from developmental exposure to an endocrine disrupting chemical to the development of adult-onset cancer. These findings provide an explanation for how human cancers, which are often associated with abnormal activation of PI3K/AKT signaling, could result from exposure to environmental insults during development.


Subject(s)
Adenocarcinoma , Phosphatidylinositol 3-Kinases , Animals , Female , Mice , Adenocarcinoma/chemically induced , beta Catenin/genetics , beta Catenin/metabolism , Cell Differentiation , Estrogens , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Uterus
3.
Dev Cell ; 53(5): 545-560.e7, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32442396

ABSTRACT

Embryonic genome activation (EGA) is orchestrated by an intrinsic developmental program initiated during oocyte maturation with translation of stored maternal mRNAs. Here, we show that tankyrase, a poly(ADP-ribosyl) polymerase that regulates ß-catenin levels, undergoes programmed translation during oocyte maturation and serves an essential role in mouse EGA. Newly translated TNKS triggers proteasomal degradation of axin, reducing targeted destruction of ß-catenin and promoting ß-catenin-mediated transcription of target genes, including Myc. MYC mediates ribosomal RNA transcription in 2-cell embryos, supporting global protein synthesis. Suppression of tankyrase activity using knockdown or chemical inhibition causes loss of nuclear ß-catenin and global reductions in transcription and histone H3 acetylation. Chromatin and transcriptional profiling indicate that development arrests prior to the mid-2-cell stage, mediated in part by reductions in ß-catenin and MYC. These findings indicate that post-transcriptional regulation of tankyrase serves as a ligand-independent developmental mechanism for post-translational ß-catenin activation and is required to complete EGA.


Subject(s)
Blastocyst/metabolism , Gene Expression Regulation, Developmental , Tankyrases/metabolism , beta Catenin/genetics , Animals , Blastocyst/cytology , Histones/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Oocytes/cytology , Oocytes/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Tankyrases/genetics , Up-Regulation , beta Catenin/metabolism
4.
Environ Health Perspect ; 128(3): 37001, 2020 03.
Article in English | MEDLINE | ID: mdl-32186404

ABSTRACT

BACKGROUND: Embryo implantation relies on precise hormonal regulation, associated gene expression changes, and appropriate female reproductive tract tissue architecture. Female mice exposed neonatally to the phytoestrogen genistein (GEN) at doses similar to those in infants consuming soy-based infant formulas are infertile due in part to uterine implantation defects. OBJECTIVES: Our goal was to determine the mechanisms by which neonatal GEN exposure causes implantation defects. METHODS: Female mice were exposed to GEN on postnatal days (PND)1-5 and uterine tissues collected on PND5, PND22-26, and during pregnancy. Analysis of tissue weights, morphology, and gene expression was performed using standard histology, confocal imaging with three-dimensional analysis, real-time reverse transcription polymerase chain reaction (real-time RT-PCR), and microarrays. The response of ovariectomized adults to 17ß-estradiol (E2) and artificial decidualization were measured. Leukemia inhibitory factor (LIF) injections were given intraperitoneally and implantation sites visualized. Gene expression patterns were compared with curated data sets to identify upstream regulators. RESULTS: GEN-exposed mice exhibited reduced uterine weight gain in response to E2 treatment or artificial decidualization compared with controls; however, expression of select hormone responsive genes remained similar between the two groups. Uteri from pregnant GEN-exposed mice were posteriorized and had reduced glandular epithelium. Implantation failure was not rescued by LIF administration. Microarray analysis of GEN-exposed uteri during early pregnancy revealed significant overlap with several conditional uterine knockout mouse models, including Foxa2, Wnt4, and Sox17. These models exhibit reduced endometrial glands, features of posteriorization and implantation failure. Expression of Foxa2, Wnt4, and Sox17, as well as genes important for neonatal uterine differentiation (Wnt7a, Hoxa10, and Msx2), were severely disrupted on PND5 in GEN-exposed mice. DISCUSSION: Our findings suggest that neonatal GEN exposure in mice disrupts expression of genes important for uterine development, causing posteriorization and diminished gland function during pregnancy that contribute to implantation failure. These findings could have implications for women who consumed soy-based formulas as infants. https://doi.org/10.1289/EHP6336.


Subject(s)
Embryo Implantation/drug effects , Genistein/adverse effects , Phytoestrogens/adverse effects , Uterus/drug effects , Animals , Female , Mice , Pregnancy , Uterus/growth & development , Uterus/physiopathology
5.
Nucleic Acids Res ; 46(11): 5487-5503, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29648668

ABSTRACT

Little is known regarding how steroid hormone exposures impact the epigenetic landscape in a living organism. Here, we took a global approach to understanding how exposure to the estrogenic chemical, diethylstilbestrol (DES), affects the neonatal mouse uterine epigenome. Integration of RNA- and ChIP-sequencing data demonstrated that ∼80% of DES-altered genes had higher H3K4me1/H3K27ac signal in close proximity. Active enhancers, of which ∼3% were super-enhancers, had a high density of estrogen receptor alpha (ERα) binding sites and were correlated with alterations in nearby gene expression. Conditional uterine deletion of ERα, but not the pioneer transcription factors FOXA2 or FOXO1, prevented the majority of DES-mediated changes in gene expression and H3K27ac signal at target enhancers. An ERα dependent super-enhancer was located at the Padi gene locus and a topological connection to the Padi1 TSS was documented using 3C-PCR. Chromosome looping at this site was independent of ERα and DES exposure, indicating that the interaction is established prior to ligand signaling. However, enrichment of H3K27ac and transcriptional activation at this locus was both DES and ERα-dependent. These data suggest that DES alters uterine development and consequently adult reproductive function by modifying the enhancer landscape at ERα binding sites near estrogen-regulated genes.


Subject(s)
Diethylstilbestrol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens, Non-Steroidal/pharmacology , Gene Expression Regulation/genetics , Regulatory Sequences, Nucleic Acid/genetics , Uterus/embryology , Animals , Binding Sites/genetics , Estrogen Receptor alpha/genetics , Estrogens, Non-Steroidal/metabolism , Female , Forkhead Box Protein O1/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Methylation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Promoter Regions, Genetic/genetics
6.
Biol Reprod ; 98(4): 449-464, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29325037

ABSTRACT

Understanding factors that regulate zygotic genome activation (ZGA) is critical for determining how cells are reprogrammed to become totipotent or pluripotent. There is limited information regarding how this process occurs physiologically in early mammalian embryos. Here, we identify a mediator complex subunit, MED13, as translated during mouse oocyte maturation and transcribed early from the zygotic genome. Knockdown and conditional knockout approaches demonstrate that MED13 is essential for ZGA in the mouse, in part by regulating expression of the embryo-specific chromatin remodeling complex, esBAF. The role of MED13 in ZGA is mediated in part by interactions with E2F transcription factors. In addition to MED13, its paralog, MED13L, is required for successful preimplantation embryo development. MED13L partially compensates for loss of MED13 function in preimplantation knockout embryos, but postimplantation development is not rescued by MED13L. Our data demonstrate an essential role for MED13 in supporting chromatin reprogramming and directed transcription of essential genes during ZGA.


Subject(s)
Embryonic Development/physiology , Gene Expression Regulation, Developmental , Mediator Complex/metabolism , Oocytes/metabolism , Animals , Chromatin/metabolism , Female , Gene Knockdown Techniques , Genome , Mediator Complex/genetics , Mice , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism
7.
EMBO J ; 36(21): 3175-3193, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29021282

ABSTRACT

Methionine metabolism is critical for epigenetic maintenance, redox homeostasis, and animal development. However, the regulation of methionine metabolism remains unclear. Here, we provide evidence that SIRT1, the most conserved mammalian NAD+-dependent protein deacetylase, is critically involved in modulating methionine metabolism, thereby impacting maintenance of mouse embryonic stem cells (mESCs) and subsequent embryogenesis. We demonstrate that SIRT1-deficient mESCs are hypersensitive to methionine restriction/depletion-induced differentiation and apoptosis, primarily due to a reduced conversion of methionine to S-adenosylmethionine. This reduction markedly decreases methylation levels of histones, resulting in dramatic alterations in gene expression profiles. Mechanistically, we discover that the enzyme converting methionine to S-adenosylmethionine in mESCs, methionine adenosyltransferase 2a (MAT2a), is under control of Myc and SIRT1. Consistently, SIRT1 KO embryos display reduced Mat2a expression and histone methylation and are sensitive to maternal methionine restriction-induced lethality, whereas maternal methionine supplementation increases the survival of SIRT1 KO newborn mice. Our findings uncover a novel regulatory mechanism for methionine metabolism and highlight the importance of methionine metabolism in SIRT1-mediated mESC maintenance and embryonic development.


Subject(s)
Embryonic Development/genetics , Epigenesis, Genetic , Methionine Adenosyltransferase/genetics , Methionine/metabolism , Mouse Embryonic Stem Cells/metabolism , Sirtuin 1/genetics , Acetylation , Animals , Apoptosis , Cell Differentiation , Embryo, Mammalian , Histones/genetics , Histones/metabolism , Metabolomics , Methionine/administration & dosage , Methionine Adenosyltransferase/metabolism , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Mouse Embryonic Stem Cells/cytology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , S-Adenosylmethionine/metabolism , Sirtuin 1/deficiency
8.
J Pathol ; 242(2): 246-259, 2017 06.
Article in English | MEDLINE | ID: mdl-28295343

ABSTRACT

Epithelial membrane protein-2 (EMP2) is a tetraspan protein predicted to regulate placental development. Highly expressed in secretory endometrium and trophectoderm cells, previous studies suggest that it may regulate implantation by orchestrating the surface expression of integrins and other membrane proteins. In order to test the role of EMP2 in pregnancy, mice lacking EMP2 (Emp2-/- ) were generated. Emp2-/- females are fertile but have reduced litter sizes when carrying Emp2-/- but not Emp2+/- fetuses. Placentas of Emp2-/- fetuses exhibit dysregulation in pathways related to neoangiogenesis, coagulation, and oxidative stress, and have increased fibrin deposition and altered vasculature. Given that these findings often occur due to placental insufficiency resulting in an oxygen-poor environment, the expression of hypoxia-inducible factor-1 alpha (HIF-1α) was examined. Placentas from Emp2-/- fetuses had increased total HIF-1α expression in large part through an increase in uterine NK (uNK) cells, demonstrating a unique interplay between uNK cells and trophoblasts modulated through EMP2. To determine if these results translated to human pregnancy, placentas from normal, term deliveries or those complicated by placental insufficiency resulting in intrauterine growth restriction (IUGR) were stained for EMP2. EMP2 was significantly reduced in both villous and extravillous trophoblast populations in IUGR placentas. Experiments in vitro using human trophoblast cells lines indicate that EMP2 modulates angiogenesis by altering HIF-1α expression. Our results reveal a novel role for EMP2 in regulating trophoblast function and vascular development in mice and humans, and suggest that it may be a new biomarker for placental insufficiency. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Fetal Growth Retardation/genetics , Membrane Glycoproteins/genetics , Oxygen/metabolism , Placental Insufficiency/genetics , Animals , Disease Models, Animal , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Fibrin/genetics , Fibrin/metabolism , Gene Knockout Techniques , Homologous Recombination , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Placenta/blood supply , Placenta/metabolism , Placenta/pathology , Placental Insufficiency/metabolism , Placental Insufficiency/pathology , Placentation , Pregnancy , Trophoblasts/metabolism , Trophoblasts/pathology , Uterus/blood supply , Uterus/metabolism , Uterus/pathology
9.
Cell Calcium ; 65: 63-72, 2017 07.
Article in English | MEDLINE | ID: mdl-28222911

ABSTRACT

Repetitive oscillations in cytoplasmic Ca2+ due to periodic Ca2+ release from the endoplasmic reticulum (ER) drive mammalian embryo development following fertilization. Influx of extracellular Ca2+ to support the refilling of ER stores is required for sustained Ca2+ oscillations, but the mechanisms underlying this Ca2+ influx are controversial. Although store-operated Ca2+ entry (SOCE) is an appealing candidate mechanism, several groups have arrived at contradictory conclusions regarding the importance of SOCE in oocytes and eggs. To definitively address this question, Ca2+ influx was assessed in oocytes and eggs lacking the major components of SOCE, the ER Ca2+ sensor STIM proteins, and the plasma membrane Ca2+ channel ORAI1. We generated oocyte-specific conditional knockout (cKO) mice for Stim1 and Stim2, and also generated Stim1/2 double cKO mice. Females lacking one or both STIM proteins were fertile and their ovulated eggs displayed normal patterns of Ca2+ oscillations following fertilization. In addition, no impairment was observed in ER Ca2+ stores or Ca2+ influx following store depletion. Similar studies were performed on eggs from mice globally lacking ORAI1; no abnormalities were observed. Furthermore, spontaneous Ca2+ influx was normal in oocytes from Stim1/2 cKO and ORAI1-null mice. Finally, we tested if TRPM7-like channels could support spontaneous Ca2+ influx, and found that it was largely prevented by NS8593, a TRPM7-specific inhibitor. Fertilization-induced Ca2+ oscillations were also impaired by NS8593. Combined, these data robustly show that SOCE is not required to support appropriate Ca2+ signaling in mouse oocytes and eggs, and that TRPM7-like channels may contribute to Ca2+ influx that was previously attributed to SOCE.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Fertilization/physiology , Oocytes/metabolism , Animals , Cytoplasm/genetics , Cytoplasm/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Mice , Mice, Knockout , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Oocytes/cytology , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/genetics , Stromal Interaction Molecule 2/metabolism
10.
Mol Cancer Res ; 14(9): 849-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27259717

ABSTRACT

UNLABELLED: The oncofetal protein sine oculis-related homeobox 1 (SIX1) is a developmental transcription factor associated with carcinogenesis in several human cancer types but has not been investigated in human endometrial cancer. In a model of hormonal carcinogenesis, mice neonatally exposed to the soy phytoestrogen genistein (GEN) or the synthetic estrogen diethylstilbestrol (DES) develop endometrial cancer as adults. Previously, we demonstrated that SIX1 becomes aberrantly expressed in the uteri of these mice. Here, we used this mouse model to investigate the role of SIX1 expression in endometrial carcinoma development and used human tissue microarrays to explore the utility of SIX1 as a biomarker in human endometrial cancer. In mice neonatally exposed to GEN or DES, the Six1 transcript level increased dramatically over time in uteri at 6, 12, and 18 months of age and was associated with development of endometrial carcinoma. SIX1 protein localized within abnormal basal cells and all atypical hyperplastic and neoplastic lesions. These findings indicate that developmental estrogenic chemical exposure induces persistent endometrial SIX1 expression that is strongly associated with abnormal cell differentiation and cancer development. In human endometrial tissue specimens, SIX1 was not present in normal endometrium but was expressed in a subset of endometrial cancers in patients who were also more likely to have late-stage disease. These findings identify SIX1 as a disease biomarker in a model of hormonal carcinogenesis and suggest that SIX1 plays a role in endometrial cancer development in both mice and women. IMPLICATIONS: The SIX1 oncoprotein is aberrantly expressed in the endometrium following developmental exposure to estrogenic chemicals, correlates with uterine cancer, and is a biomarker in human endometrial cancers. Mol Cancer Res; 14(9); 849-58. ©2016 AACR.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinogenesis/metabolism , Endometrial Neoplasms/metabolism , Homeodomain Proteins/biosynthesis , Animals , Animals, Newborn , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Differentiation/physiology , Diethylstilbestrol , Disease Models, Animal , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Genistein , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Transcription Factors/metabolism , Uterus/metabolism , Uterus/pathology
11.
Elife ; 4: e10453, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26623518

ABSTRACT

Development of uterine endometrial receptivity for implantation is orchestrated by cyclic steroid hormone-mediated signals. It is unknown if these signals are necessary for oviduct function in supporting fertilization and preimplantation development. Here we show that conditional knockout (cKO) mice lacking estrogen receptor α (ERα) in oviduct and uterine epithelial cells have impaired fertilization due to a dramatic reduction in sperm migration. In addition, all successfully fertilized eggs die before the 2-cell stage due to persistence of secreted innate immune mediators including proteases. Elevated protease activity in cKO oviducts causes premature degradation of the zona pellucida and embryo lysis, and wild-type embryos transferred into cKO oviducts fail to develop normally unless rescued by concomitant transfer of protease inhibitors. Thus, suppression of oviductal protease activity mediated by estrogen-epithelial ERα signaling is required for fertilization and preimplantation embryo development. These findings have implications for human infertility and post-coital contraception.


Subject(s)
Estrogen Receptor alpha/agonists , Estrogens/metabolism , Fertilization , Oviducts/physiology , Peptide Hydrolases/metabolism , Signal Transduction , Animals , Embryo Loss , Estrogen Receptor alpha/genetics , Female , Gene Knockdown Techniques , Mice , Oviducts/drug effects , Uterus/drug effects , Uterus/physiology
12.
J Cell Sci ; 128(23): 4442-52, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26483387

ABSTRACT

Initiation of mouse embryonic development depends upon a series of fertilization-induced rises in intracellular Ca(2+). Complete egg activation requires influx of extracellular Ca(2+); however, the channels that mediate this influx remain unknown. Here, we tested whether the α1 subunit of the T-type channel CaV3.2, encoded by Cacna1h, mediates Ca(2+) entry into oocytes. We show that mouse eggs express a robust voltage-activated Ca(2+) current that is completely absent in Cacna1h(-/-) eggs. Cacna1h(-/-) females have reduced litter sizes, and careful analysis of Ca(2+) oscillation patterns in Cacna1h(-/-) eggs following in vitro fertilization (IVF) revealed reductions in first transient length and oscillation persistence. Total and endoplasmic reticulum (ER) Ca(2+) stores were also reduced in Cacna1h(-/-) eggs. Pharmacological inhibition of CaV3.2 in wild-type CF-1 strain eggs using mibefradil or pimozide reduced Ca(2+) store accumulation during oocyte maturation and reduced Ca(2+) oscillation persistence, frequency and number following IVF. Overall, these data show that CaV3.2 T-type channels have prev8iously unrecognized roles in supporting the meiotic-maturation-associated increase in ER Ca(2+) stores and mediating Ca(2+) influx required for the activation of development.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Fertilization/physiology , Oocytes/metabolism , Animals , Calcium Channels, T-Type/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Female , Mice , Mice, Knockout , Oocytes/cytology
13.
Development ; 142(15): 2633-40, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26160904

ABSTRACT

During oocyte maturation, capacity and sensitivity of Ca(2+) signaling machinery increases dramatically, preparing the metaphase II (MII)-arrested egg for fertilization. Upon sperm-egg fusion, Ca(2+) release from IP3-sensitive endoplasmic reticulum stores results in cytoplasmic Ca(2+) oscillations that drive egg activation and initiate early embryo development. Premature Ca(2+) release can cause parthenogenetic activation prior to fertilization; thus, preventing inappropriate Ca(2+) signaling is crucial for ensuring robust MII arrest. Here, we show that regulator of G-protein signaling 2 (RGS2) suppresses Ca(2+) release in MII eggs. Rgs2 mRNA was recruited for translation during oocyte maturation, resulting in ∼ 20-fold more RGS2 protein in MII eggs than in fully grown immature oocytes. Rgs2-siRNA-injected oocytes matured to MII; however, they had increased sensitivity to low pH and acetylcholine (ACh), which caused inappropriate Ca(2+) release and premature egg activation. When matured in vitro, RGS2-depleted eggs underwent spontaneous Ca(2+) increases that were sufficient to cause premature zona pellucida conversion. Rgs2(-/-) females had reduced litter sizes, and their eggs had increased sensitivity to low pH and ACh. Rgs2(-/-) eggs also underwent premature zona pellucida conversion in vivo. These findings indicate that RGS2 functions as a brake to suppress premature Ca(2+) release in eggs that are poised on the brink of development.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Ovum/physiology , RGS Proteins/metabolism , Sperm-Ovum Interactions/physiology , Animals , Female , Fluorescent Antibody Technique , Immunoblotting , Mice , Ovum/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
14.
Biol Reprod ; 90(3): 63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24501176

ABSTRACT

Fully grown oocytes in the ovary are arrested at prophase of meiosis I because of high levels of intraoocyte cAMP that maintain increased levels of cAMP-dependent protein kinase (PKA) activity. Following the luteinizing hormone surge at the time of ovulation, cAMP levels drop, resulting in a reduction in PKA activity that triggers meiotic resumption. Although much is known about the molecular mechanisms of how PKA activity fluctuations initiate the oocyte's reentry into meiosis, significantly less is known about the requirement for PKA activity in the oocyte after exit from the prophase I arrest. Here we show that although PKA activity decreases in the oocyte upon meiotic resumption, it increases throughout meiotic progression from the time of germinal vesicle breakdown (GVBD) until the metaphase II (MII) arrest. Blocking this meiotic maturation-associated increase in PKA activity using the pharmacological inhibitor H89 resulted in altered kinetics of GVBD, defects in chromatin and spindle dynamics, and decreased ability of oocytes to reach MII. These effects appear to be largely PKA specific because inhibitors targeting other kinases did not have the same outcomes. To determine potential proteins that may require PKA phosphorylation during meiosis, we separated oocyte protein extracts on an SDS-PAGE gel, extracted regions of the gel that had corresponding immune reactivity towards an anti-PKA substrate antibody, and performed mass spectrometry and microsequencing. Using this approach, we identified transducin-like enhancer of split-6 (TLE6)-a maternal effect gene that is part of the subcortical maternal complex-as a putative PKA substrate. TLE6 localized to the oocyte cortex throughout meiosis in a manner that is spatially and temporally consistent with the localization of critical PKA subunits. Moreover, we demonstrated that TLE6 becomes phosphorylated in a narrow window following meiotic resumption, and H89 treatment can completely block this phosphorylation when added prior to GVBD but not after. Taken together, these results highlight the importance of oocyte-intrinsic PKA in regulating meiotic progression after the prophase I arrest and offer new insights into downstream targets of its activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Oocytes/physiology , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Co-Repressor Proteins , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Immunoprecipitation , Isoquinolines/metabolism , Isoquinolines/pharmacology , Male , Mass Spectrometry , Meiosis/physiology , Meiotic Prophase I/drug effects , Metaphase/physiology , Mice , Molecular Sequence Data , Oligopeptides/metabolism , Oocytes/enzymology , Oocytes/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Substrate Specificity , Sulfonamides/metabolism , Sulfonamides/pharmacology
15.
PLoS One ; 9(1): e85894, 2014.
Article in English | MEDLINE | ID: mdl-24465770

ABSTRACT

Bisphenol A (BPA) exposure results in numerous developmental and functional abnormalities in reproductive organs in rodent models, but limited data are available regarding BPA effects in the primate uterus. To determine if maternal oral BPA exposure affects fetal uterine development in a non-human primate model, pregnant rhesus macaques carrying female fetuses were exposed orally to 400 µg/kg BPA or vehicle control daily from gestation day (GD) 50-100 or GD100-165. Fetal uteri were collected at the completion of treatment (GD100 or GD165); tissue histology, cell proliferation, and expression of estrogen receptor alpha (ERα) and progesterone receptor (PR) were compared to that of controls. Gene expression analysis was conducted using rhesus macaque microarrays. There were no significant differences in histology or in the percentage of cells expressing the proliferation marker Ki-67, ERα, or PR in BPA-exposed uteri compared to controls at GD100 or GD165. Minimal differences in gene expression were observed between BPA-exposed and control GD100 uteri. However, at GD165, BPA-exposed uteri had significant differences in gene expression compared to controls. Several of the altered genes, including HOXA13, WNT4, and WNT5A, are critical for reproductive organ development and/or adult function. We conclude that second or third trimester BPA exposure does not significantly affect fetal uterus development based on morphological, proliferation, and steroid hormone receptor assessments. However, differences in expression of key developmental genes after third trimester exposure suggest that BPA could alter transcriptional signals influencing uterine function later in life.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Gene Expression Regulation, Developmental/drug effects , Phenols/toxicity , Uterus/drug effects , Animals , Environmental Exposure , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Fetal Development/drug effects , Gene Regulatory Networks , Macaca mulatta , Maternal Exposure , Maternal-Fetal Exchange , Oligonucleotide Array Sequence Analysis , Pregnancy , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Transcriptome/drug effects , Uterus/embryology , Uterus/metabolism , Uterus/pathology , beta Catenin/metabolism
16.
Mol Endocrinol ; 27(10): 1666-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24002655

ABSTRACT

Neonatal exposure to diethylstilbestrol (DES) causes permanent alterations in female reproductive tract gene expression, infertility, and uterine cancer in mice. To determine whether epigenetic mechanisms could explain these phenotypes, we first tested whether DES altered uterine expression of chromatin-modifying proteins. DES treatment significantly reduced expression of methylcytosine dioxygenase TET oncogene family, member 1 (TET1) on postnatal day 5; this decrease was correlated with a subtle decrease in DNA 5-hydroxymethylcytosine in adults. There were also significant reductions in histone methyltransferase enhancer of zeste homolog 2 (EZH2), histone lysine acetyltransferase 2A (KAT2A), and histone deacetylases HDAC1, HDAC2, and HDAC3. Uterine chromatin immunoprecipitation was used to analyze the locus-specific association of modified histones with 2 genes, lactoferrin (Ltf) and sine oculis homeobox 1 (Six1), which are permanently upregulated in adults after neonatal DES treatment. Three histone modifications associated with active transcription, histone H3 lysine 9 acetylation (H3K9ac), H3 lysine 4 trimethylation (H3K4me3), and H4 lysine 5 acetylation (H4K5ac) were enriched at specific Ltf promoter regions after DES treatment, but this enrichment was not maintained in adults. H3K9ac, H4K5ac, and H3K4me3 were enriched at Six1 exon 1 immediately after neonatal DES treatment. As adults, DES-treated mice had greater differences in H4K5ac and H3K4me3 occupancy at Six1 exon 1 and new differences in these histone marks at an upstream region. These findings indicate that neonatal DES exposure temporarily alters expression of multiple chromatin-modifying proteins and persistently alters epigenetic marks in the adult uterus at the Six1 locus, suggesting a mechanism for developmental exposures leading to altered reproductive function and increased cancer risk.


Subject(s)
Diethylstilbestrol/pharmacology , Epigenesis, Genetic/drug effects , Estrogens, Non-Steroidal/pharmacology , Uterus/drug effects , Animals , Animals, Newborn , DNA-Cytosine Methylases/genetics , DNA-Cytosine Methylases/metabolism , Endocrine Disruptors/pharmacology , Female , Gene Expression , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Methylation , Mice , Protein Processing, Post-Translational , Uterus/pathology
17.
J Am Assoc Lab Anim Sci ; 52(2): 130-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23562095

ABSTRACT

The lowest observed adverse effect level for bisphenol A (BPA) in mice and rats is currently poorly defined due to inconsistent study designs and results in published studies. The objectives of the current study were to (1) compare the estrogenic content of rodent diets, bedding, cages, and water bottles to evaluate their impact on the estrogenic activity of BPA and (2) review the literature on BPA to determine the most frequently reported diets, beddings, cages, and water bottles used in animal studies. Our literature review indicated that low-dose BPA animal studies have inconsistent results and that factors contributing to this inconsistency are the uses of high-phytoestrogen diets and the different routes of exposure. In 44% (76 of 172) of all reports, rodents were exposed to BPA via the subcutaneous route. Our literature review further indicated that the type of diet, bedding, caging, and water bottles used in BPA studies were not always reported. Only 37% (64 of 172) of the reports described the diet used. In light of these findings, we recommend the use of a diet containing low levels of phytoestrogen (less than 20 µg/g diet) and metabolizable energy (approximately 3.1 kcal/g diet) and estrogen-free bedding, cages, and water bottles for studies evaluating the estrogenic activity of endocrine-disrupting compounds such as BPA. The oral route of BPA exposure should be used when results are to be extrapolated to humans.


Subject(s)
Benzhydryl Compounds/adverse effects , Benzhydryl Compounds/analysis , Endocrine Disruptors/adverse effects , Phenols/adverse effects , Phenols/analysis , Animal Feed/analysis , Animals , Endocrine Disruptors/analysis , Energy Metabolism , Female , Housing, Animal , Male , Mice , Phytoestrogens/adverse effects , Phytoestrogens/analysis , Rats
18.
Biol Reprod ; 87(1): 10, 1-10, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553218

ABSTRACT

Treatment of neonatal mice with the phytoestrogen genistein (50 mg/kg/day) results in complete female infertility caused in part by preimplantation embryo loss in the oviduct between Days 2 and 3 of pregnancy. We previously demonstrated that oviducts of genistein-treated mice are "posteriorized" as compared to control mouse oviducts because they express numerous genes normally restricted to posterior regions of the female reproductive tract (FRT), the cervix and vagina. We report here that neonatal genistein treatment resulted in substantial changes in oviduct expression of genes important for the FRT mucosal immune response, including immunoglobulins, antimicrobials, and chemokines. Some of the altered immune response genes were chronically altered beginning at the time of neonatal genistein treatment, indicating that these alterations were a result of the posteriorization phenotype. Other alterations in oviduct gene expression were observed only in early pregnancy, immediately after the FRT was exposed to inflammatory or antigenic stimuli from ovulation and mating. The oviduct changes affected development of the surviving embryos by increasing the rate of cleavage and decreasing the trophectoderm-to-inner cell mass cell ratio at the blastocyst stage. We conclude that both altered immune responses to pregnancy and deficits in oviduct support for preimplantation embryo development in the neonatal genistein model are likely to contribute to infertility phenotype.


Subject(s)
Embryonic Development/drug effects , Genistein/toxicity , Immunity, Mucosal/drug effects , Oviducts/drug effects , Oviducts/immunology , Phytoestrogens/toxicity , Animals , Animals, Newborn , Embryonic Development/genetics , Embryonic Development/immunology , Embryonic Development/physiology , Female , Gene Expression Regulation, Developmental/drug effects , Genes, MHC Class II/drug effects , Genistein/administration & dosage , Immunity, Mucosal/genetics , Infertility, Female/chemically induced , Infertility, Female/genetics , Infertility, Female/immunology , Infertility, Female/metabolism , Inflammation Mediators/metabolism , Male , Mice , Oviducts/metabolism , Oviducts/pathology , Phytoestrogens/administration & dosage , Pregnancy
19.
Proc Natl Acad Sci U S A ; 109(11): 4169-74, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22371584

ABSTRACT

Mammalian fertilization is accompanied by oscillations in egg cytoplasmic calcium (Ca(2+)) concentrations that are critical for completion of egg activation. These oscillations are initiated by Ca(2+) release from inositol 1,4,5-trisphosphate (IP(3))-sensitive intracellular stores. We tested the hypothesis that Ca(2+) influx across the plasma membrane was a requisite component of egg activation signaling, and not simply a Ca(2+) source for store repletion. Using intracytoplasmic sperm injection (ICSI) and standard in vitro fertilization (IVF), we found that Ca(2+) influx was not required to initiate resumption of meiosis II. However, even if multiple oscillations in intracellular Ca(2+) occurred, in the absence of Ca(2+) influx, the fertilized eggs failed to emit the second polar body, resulting in formation of three pronuclei. Additional experiments using the Ca(2+) chelator, BAPTA/AM, demonstrated that Ca(2+) influx is sufficient to support polar body emission and pronucleus formation after only a single sperm-induced Ca(2+) transient, whereas BAPTA/AM-treated ICSI or fertilized eggs cultured in Ca(2+)-free medium remained arrested in metaphase II. Inhibition of store-operated Ca(2+) entry had no effect on ICSI-induced egg activation, so Ca(2+) influx through alternative channels must participate in egg activation signaling. Ca(2+) influx appears to be upstream of CaMKIIγ activity because eggs can be parthenogenetically activated with a constitutively active form of CaMKIIγ in the absence of extracellular Ca(2+). These results suggest that Ca(2+) influx at fertilization not only maintains Ca(2+) oscillations by replenishing Ca(2+) stores, but also activates critical signaling pathways upstream of CaMKIIγ that are required for second polar body emission.


Subject(s)
Calcium Signaling , Calcium/metabolism , Ovum/cytology , Ovum/metabolism , Animals , Buffers , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Cycle/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chromatin/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Extracellular Space/drug effects , Extracellular Space/metabolism , Fertilization in Vitro , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Models, Biological , Ovum/drug effects , Sperm Injections, Intracytoplasmic
SELECTION OF CITATIONS
SEARCH DETAIL