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










Publication year range
1.
Biol Reprod ; 92(1): 23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25376232

ABSTRACT

Cumulus cells and mural granulosa cells (MGCs) have functionally distinct roles in antral follicles, and comparison of their transcriptomes at a global and systems level can propel future studies on mechanisms underlying their functional diversity. These cells were isolated from small and large antral follicles before and after stimulation of immature mice with gonadotropins, respectively. Both cell types underwent dramatic transcriptomic changes, and differences between them increased with follicular growth. Although cumulus cells of both stages of follicular development are competent to undergo expansion in vitro, they were otherwise remarkably dissimilar with transcriptomic changes quantitatively equivalent to those of MGCs. Gene ontology analysis revealed that cumulus cells of small follicles were enriched in transcripts generally associated with catalytic components of metabolic processes, while those from large follicles were involved in regulation of metabolism, cell differentiation, and adhesion. Contrast of cumulus cells versus MGCs revealed that cumulus cells were enriched in transcripts associated with metabolism and cell proliferation while MGCs were enriched for transcripts involved in cell signaling and differentiation. In vitro and in vivo models were used to test the hypothesis that higher levels of transcripts in cumulus cells versus MGCs is the result of stimulation by oocyte-derived paracrine factors (ODPFs). Surprisingly ∼48% of transcripts higher in cumulus cells than MGCs were not stimulated by ODPFs. Those stimulated by ODPFs were mainly associated with cell division, mRNA processing, or the catalytic pathways of metabolism, while those not stimulated by ODPFs were associated with regulatory processes such as signaling, transcription, phosphorylation, or the regulation of metabolism.


Subject(s)
Cumulus Cells/metabolism , Granulosa Cells/metabolism , Transcriptome , Algorithms , Animals , Cells, Cultured , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Microarray Analysis , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism
2.
Biol Reprod ; 91(6): 142, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253739

ABSTRACT

Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are oocyte-secreted paralogs of the transforming growth factor beta (TGFbeta) superfamily. In mammals, these two growth factors play critical roles in folliculogenesis. As previously reported, an arginine in the pre-helix loop of GDF5 defines the high binding specificity to its type 1 receptor. Interestingly, bioactive mouse GDF9 and human BMP15 share the conserved arginine in the pre-helix loop, but their low-activity counterparts (mouse BMP15 and human GDF9) have a glycine or a proline instead. To address the question of whether the arginine residue defines the different activities of GDF9 and BMP15 homodimers and their heterodimers in human and mouse, we used site-directed mutagenesis to change the species-specific residues in human and mouse proteins, and examined their activities in our in vitro assays. Although amino acid 72 of mature GDF9 is responsible for altered homodimer bioactivities, neither the corresponding BMP15 amino acid 62 nor the intact pre-helix loop is indispensable for BMP15 homodimer activity. However, amino acid 72 in GDF9 only has only subtle effects on GDF9:BMP15 heterodimer activity. Based on previous studies and our recent findings, we provide hypothetical models to understand the molecular mechanism to define activities of the homodimeric and heterodimeric ligands. The arginine residue in the pre-helix loop of GDF9 homodimer may prevent the inhibition from its pro-domain or directly alter receptor binding, but this residue in GDF9 does not significantly affect the heterodimer activity, because of suggested conformational changes during heterodimer formation.


Subject(s)
Bone Morphogenetic Protein 15/metabolism , Growth Differentiation Factor 9 , Protein Interaction Domains and Motifs , Protein Multimerization , Amino Acid Sequence , Animals , Cells, Cultured , Female , Growth Differentiation Factor 9/chemistry , Growth Differentiation Factor 9/genetics , Growth Differentiation Factor 9/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Multimerization/genetics , Protein Structure, Secondary
3.
Endocrinology ; 154(12): 4859-72, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24035995

ABSTRACT

Oocyte-derived paracrine factors (ODPFs) and estrogens are both essential for the development and function of ovarian follicles in mammals. Cooperation of these two factors was assessed in vitro using intact cumulus-oocyte complexes, cumulus cells cultured after the removal of oocytes [oocytectomized (OOX) cumulus cells], and OOX cumulus cells cocultured with denuded oocytes, all in the presence or absence of 17ß-estradiol (E2). Effects on the cumulus cell transcriptome were assessed by microarray analysis. There was no significant difference between the cumulus cell transcriptomes of either OOX cumulus cells cocultured with oocytes or intact cumulus-oocyte complexes. Therefore, oocyte-mediated regulation of the cumulus cell transcriptome is mediated primarily by ODPFs and not by gap junctional communication between oocytes and cumulus cells. Gene ontology analysis revealed that both ODPFs and E2 strongly affected the biological processes associated with cell proliferation in cumulus cells. E2 had limited effects on ODPF-regulated biological processes. However, in sharp contrast, ODPFs significantly affected biological processes regulated by E2 in cumulus cells. For example, only in the presence of ODPFs did E2 significantly promote the biological processes related to phosphorylation-mediated signal transduction in cumulus cells, such as the signaling pathways of epidermal growth factor, vascular endothelial growth factor, and platelet-derived growth factor. Therefore, ODPFs and E2 cooperate to regulate the cumulus cell transcriptome and, in general, oocytes modulate the effects of estrogens on cumulus cell function.


Subject(s)
Cumulus Cells/metabolism , Estradiol/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Oocytes/metabolism , Transcriptome/physiology , Animals , Female , Gene Expression Regulation/drug effects , Mice , Protein Array Analysis
5.
Proc Natl Acad Sci U S A ; 110(39): E3723-9, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23980176

ABSTRACT

Coordinated regulation of oocyte and ovarian follicular development is essential for fertility. In particular, the progression of meiosis, a germ cell-specific cell division that reduces the number of chromosomes from diploid to haploid, must be arrested until just before ovulation. Follicular somatic cells are well-known to impose this arrest, which is essential for oocyte-follicle developmental synchrony. Follicular somatic cells sustain meiotic arrest via the natriuretic peptide C/natriuretic peptide receptor 2 (NPPC/NPR2) system, and possibly also via high levels of the purine hypoxanthine in the follicular fluid. Upon activation by the ligand NPPC, NPR2, the predominant guanylyl cyclase in follicular somatic cells, produces cyclic guanosine monophosphate (cGMP), which maintains meiotic arrest after transfer to the oocyte via gap junctions. Here we report that both the NPPC/NPR2 system and hypoxanthine require the activity of inosine monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme required for the production of guanylyl metabolites and cGMP. Furthermore, oocyte-derived paracrine factors, particularly the growth differentiation factor 9-bone morphogenetic protein 15 heterodimer, promote expression of Impdh and Npr2 and elevate cGMP levels in cumulus cells. Thus, although the somatic compartment of ovarian follicles plays an essential role in the maintenance of oocyte meiotic arrest, as has been known for many years, this function of the somatic cells is surprisingly regulated by signals from the oocyte itself.


Subject(s)
Cell Communication , Mammals/metabolism , Meiosis , Oocytes/cytology , Ovarian Follicle/cytology , Animals , Cell Communication/genetics , Cumulus Cells/cytology , Cumulus Cells/metabolism , Cyclic GMP/biosynthesis , Female , Gene Expression Regulation, Enzymologic , Hypoxanthine/metabolism , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Meiosis/genetics , Models, Biological , Oocytes/enzymology , Paracrine Communication , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction/genetics
6.
Proc Natl Acad Sci U S A ; 110(8): E776-85, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382188

ABSTRACT

The TGF-ß superfamily is the largest family of secreted proteins in mammals, and members of the TGF-ß family are involved in most developmental and physiological processes. Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), oocyte-secreted paralogs of the TGF-ß superfamily, have been shown genetically to control ovarian physiology. Although previous studies found that GDF9 and BMP15 homodimers can modulate ovarian pathways in vitro, the functional species-specific significance of GDF9:BMP15 heterodimers remained unresolved. Therefore, we engineered and produced purified recombinant mouse and human GDF9 and BMP15 homodimers and GDF9:BMP15 heterodimers to compare their molecular characteristics and physiological functions. In mouse granulosa cell and cumulus cell expansion assays, mouse GDF9 and human BMP15 homodimers can up-regulate cumulus expansion-related genes (Ptx3, Has2, and Ptgs2) and promote cumulus expansion in vitro, whereas mouse BMP15 and human GDF9 homodimers are essentially inactive. However, we discovered that mouse GDF9:BMP15 heterodimer is ∼10- to 30-fold more biopotent than mouse GDF9 homodimer, and human GDF9:BMP15 heterodimer is ∼1,000- to 3,000-fold more bioactive than human BMP15 homodimer. We also demonstrate that the heterodimers require the kinase activities of ALK4/5/7 and BMPR2 to activate SMAD2/3 but unexpectedly need ALK6 as a coreceptor in the signaling complex in granulosa cells. Our findings that GDF9:BMP15 heterodimers are the most bioactive ligands in mice and humans compared with homodimers explain many puzzling genetic and physiological data generated during the last two decades and have important implications for improving female fertility in mammals.


Subject(s)
Bone Morphogenetic Protein 15/physiology , Growth Differentiation Factor 9/physiology , Ovary/physiology , Animals , Bone Morphogenetic Protein 15/metabolism , Female , Growth Differentiation Factor 9/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad Proteins/metabolism
7.
Biol Reprod ; 88(2): 42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23255339

ABSTRACT

Natriuretic peptide type C (NPPC) and its receptor natriuretic peptide receptor 2 (NPR2) regulate cGMP in ovarian follicles and participate in maintaining oocyte meiotic arrest. We investigated the regulation of Nppc expression in mouse granulosa cells in vivo and in vitro. In mural granulosa cells (MGCs) in vivo, eCG caused an increase in Nppc mRNA, and subsequent human chorionic gonadotropin (hCG) treatment caused a decrease. A culture system was established for MGCs isolated from follicles not stimulated with equine chorionic gonadotropin to further define the mechanisms controlling Nppc expression. In this system, expression of Nppc mRNA was increased by estradiol (E2), with augmentation by follicle-stimulating hormone (FSH), but FSH or luteinizing hormone (LH) alone had no effect. Thus, estrogens are important for regulating Nppc expression, probably by feedback mechanisms enhancing the action of gonadotropins. In MGCs treated with E2 plus FSH in vitro, subsequent treatment with EGF, but not LH, decreased Nppc mRNA. MGCs express higher levels of both Nppc and Lhcgr mRNAs than cumulus cells. Oocyte-derived paracrine factors suppressed cumulus cell Lhcgr but not Nppc expression. Thus, higher Nppc expression by MGCs is not the result of oocyte suppression of expression in cumulus cells. Another possible regulator of the LH-induced NPPC decrease is NPR3, an NPPC clearance receptor. Human chorionic gonadotropin increased Npr3 expression in vivo and LH increased Npr3 mRNA in cultured MGCs, independently of EGF receptor activation. Interestingly, despite the increase in Npr3 mRNA, the hCG-induced decrease in ovarian NPPC occurred normally in an Npr3 mutant (lgj), thus NPR3 probably does not participate in regulation of ovarian NPPC levels or oocyte development.


Subject(s)
Epidermal Growth Factor/pharmacology , Estradiol/pharmacology , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Luteinizing Hormone/pharmacology , Natriuretic Peptide, C-Type/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Cell Communication/physiology , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Cyclic GMP/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/drug effects , In Vitro Techniques , Mice , Mice, Mutant Strains , Models, Animal , Mutation/genetics , Natriuretic Peptide, C-Type/genetics , Oocytes/cytology , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/genetics
8.
Endocrinology ; 152(11): 4377-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21914782

ABSTRACT

Natriuretic peptide type C (NPPC) and its cognate receptor natriuretic peptide receptor 2 (NPR2) are essential for maintaining meiotic arrest in mouse oocytes residing in Graafian follicles. Cumulus cells, which are associated with the oocyte, express the receptor NPR2, a guanylyl cyclase, whereas mural granulosa cells express ligand NPPC. This study determined the temporal expression of Npr2 and the hormonal factors that participate in regulating its expression and, thereby, in oocyte meiotic arrest. Stimulation of follicular development in vivo with equine chorionic gonadotropin (eCG) promoted expression of Npr2 mRNA by cumulus cells and some periantral mural granulosa cells. However, FSH did not elevate the levels of Npr2 mRNA in cultured cumulus-oocyte complexes (COCs) isolated from mice not stimulated in vivo with eCG. Nevertheless, estradiol elevated expression of this transcript in vitro to the same steady-state level found in COCs isolated from eCG-stimulated follicles in vivo. Expression of Npr2 mRNA was rapidly reduced in COCs in vitro after isolation from eCG-primed mice unless maintained in culture with estradiol. The ability of NPPC to maintain meiotic arrest in cultured COCs was transient unless culture was in estradiol-containing medium. Ability of cumulus cells to produce cyclic GMP, which is required for the maintenance of meiotic arrest, was also lost in the absence of estradiol, indicating that estradiol is required to maintain functional NPR2 receptors on cumulus cells in vitro. It is concluded that estradiol promotes and maintains expression of NPR2 in cumulus cells and participates in NPPC-mediated maintenance of oocyte meiotic arrest in vitro.


Subject(s)
Cumulus Cells/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Oocytes/cytology , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism
9.
Mol Endocrinol ; 24(12): 2303-14, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21047911

ABSTRACT

The differentiation and function of cumulus cells depend upon oocyte-derived paracrine factors, but studies on the estrogen receptor knockout mice suggested that estrogen also participates in these processes. This study investigates the possible coordination of estrogen and oocytes in the development and function of cumulus cells using cumulus expansion and the expression of transcripts required for expansion as functional endpoints. Preantral granulosa cell-oocyte complexes developed in vitro with 17ß-estradiol (E2) exhibited increased levels of cumulus expansion and Has2 transcripts, encoding hyaluronan synthase 2, compared with those developed without E2. Moreover, cumulus cell-oocyte complexes (COCs) isolated from antral follicles and maintained in culture without E2 exhibited reduced cumulus expansion and Has2 mRNA levels compared with freshly isolated COCs. Exogenous E2, provided during the maintenance culture, alleviated these deficiencies. However, when oocytes were removed from COCs, E2 supplementation did not maintain competence to undergo expansion; the presence in culture of either fully grown oocytes or recombinant growth differentiation factor 9 (GDF9) was required. Recombinant bone morphogenetic protein 15, but not fibroblast growth factor 8, augmented the GDF9 effect. Oocytes or GDF9 suppressed cumulus cell levels of Nrip1 transcripts encoding nuclear receptor-interacting protein 1, a potential inhibitor of estrogen receptor signals. Therefore, E2 and oocyte-derived paracrine factors GDF9 and bone morphogenetic protein 15 coordinate to promote the development of cumulus cells and maintain their competence to undergo expansion. Furthermore, suppression of Nrip1 expression in cumulus cells by oocyte may be one mechanism mediating cross talk between oocyte and E2 signals that promotes follicular development.


Subject(s)
Bone Morphogenetic Protein 15/metabolism , Cumulus Cells/drug effects , Estradiol/pharmacology , Growth Differentiation Factor 9/metabolism , Oocytes/drug effects , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , C-Reactive Protein/genetics , Cell Adhesion Molecules/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cumulus Cells/cytology , Cumulus Cells/physiology , Cyclooxygenase 2/genetics , Estrogens/physiology , Female , Fibroblast Growth Factor 8/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Receptor Interacting Protein 1 , Oocytes/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
10.
Mol Endocrinol ; 24(6): 1230-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382892

ABSTRACT

LH triggers the maturation of the cumulus-oocyte complex (COC), which is followed by ovulation. These ovarian follicular responses to LH are mediated by epidermal growth factor (EGF)-like growth factors produced by granulosa cells and require the participation of oocyte-derived paracrine factors. However, it is not clear how oocytes coordinate with the EGF receptor (EGFR) signaling to achieve COC maturation. The aim of the present study was to test the hypothesis that oocytes promote the expression of EGFR by cumulus cells, thus enabling them to respond to the LH-induced EGF-like peptides. Egfr mRNA and protein expression were dramatically reduced in cumulus cells of mutant mice deficient in the production of the oocyte-derived paracrine factors growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15). Moreover, microsurgical removal of oocytes from wild-type COCs dramatically reduced expression of Egfr mRNA and protein, and these levels were restored by either coculture with oocytes or treatment with recombinant GDF9 or GDF9 plus recombinant BMP15. Blocking Sma- and Mad-related protein (SMAD)2/3 phosphorylation in vitro inhibited Egfr expression in wild-type COCs and in GDF9-treated wild-type cumulus cells, and conditional deletion of Smad2 and Smad3 genes in granulosa cells in vivo resulted in the reduction of Egfr mRNA in cumulus cells. These results indicate that oocytes promote expression of Egfr in cumulus cells, and a SMAD2/3-dependent pathway is involved in this process. At least two oocyte-derived growth factors, GDF9 and BMP15, are required for EGFR expression by cumulus cells.


Subject(s)
Cumulus Cells/cytology , Cumulus Cells/metabolism , ErbB Receptors/metabolism , Luteinizing Hormone/pharmacology , Oocytes/cytology , Oocytes/metabolism , Signal Transduction/drug effects , Animals , Benzamides/pharmacology , Bone Morphogenetic Protein 15/deficiency , Bone Morphogenetic Protein 15/pharmacology , Cell Separation , Chorionic Gonadotropin/pharmacology , Coculture Techniques , Cumulus Cells/drug effects , Dioxoles/pharmacology , ErbB Receptors/genetics , Female , Gene Expression Regulation/drug effects , Growth Differentiation Factor 9/deficiency , Growth Differentiation Factor 9/pharmacology , Humans , Mice , Mutation/genetics , Oocytes/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism
11.
PLoS One ; 4(10): e7479, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19834616

ABSTRACT

BACKGROUND: Gene expression microarrays have provided many insights into changes in gene expression patterns between different tissue types, developmental stages, and disease states. Analyses of these data focused primarily measuring the relative abundance of transcripts of a gene, while treating most or all transcript isoforms as equivalent. Differences in the selection between transcript isoforms can, however, represent critical changes to either the protein product or the posttranscriptional regulation of the transcript. Novel analyses on existing microarray data provide fresh insights and new interpretations into transcriptome-wide changes in expression. METHODOLOGY: A probe-level analysis of existing gene expression arrays revealed differences in mRNA processing, primarily affecting the 3'-untranslated region. Working with the example of microarrays drawn from a transcriptionally silent period of mouse oocyte development, probe-level analysis (implemented here as rmodel) identified genes whose transcript isoforms have differing stabilities. Comparison of micorarrays measuring cDNA generated from oligo-dT and random primers revealed further differences in the polyadenylation status of some transcripts. Additional analysis provided evidence for sequence-targeted cleavage, including putative targeting sequences, as one mechanism of degradation for several hundred transcripts in the maturing oocyte. CONCLUSIONS: The capability of probe-level analysis to elicit novel findings from existing expression microarray data was demonstrated. The characterization of differences in stability between transcript isoforms in maturing mouse oocytes provided some mechanistic details of degradation. Similar analysis of existing archives of expression microarray data will likely provide similar discoveries.


Subject(s)
Gene Expression Regulation , Oligonucleotide Array Sequence Analysis/methods , Oocytes/cytology , Oocytes/metabolism , Animals , DNA Probes/chemistry , DNA, Complementary/metabolism , Expressed Sequence Tags , Female , Mice , Mice, Inbred C57BL , Models, Biological , Oligonucleotide Probes/chemistry , Protein Isoforms , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
12.
Biol Reprod ; 81(5): 833-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19553596

ABSTRACT

Mouse oocytes produce members of the transforming growth factor beta (TGFbeta) superfamily, including bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9), as well as fibroblast growth factors (FGFs). These growth factors cooperate to regulate cumulus cell function. To identify potential mechanisms involved in these interactions, the ability of fully grown oocytes to regulate expression of BMP or FGF antagonists in cumulus cells was examined. Oocytes promoted cumulus cell expression of transcripts encoding antagonists to TGFbeta superfamily members, including Grem2, Htra1, Htra3, and Nog mRNAs. In contrast, oocytes suppressed cumulus cell expression of Spry2 mRNA, which encodes a regulator of receptor tyrosine kinase signals, such as FGF and epidermal growth factor (EGF) receptor signals. The regulation of Spry2 mRNA levels in cumulus cells was studied further as a model for analysis of potential mechanisms for cooperativity of FGF/EGF signaling with oocyte-derived members of the TGFbeta superfamily. Oocytes suppressed basal and FGF-stimulated Spry2 mRNA levels in cumulus cells but promoted EGF-stimulated levels. Furthermore, recombinant TGFbeta superfamily proteins, including BMP15 and GDF9, mimicked these effects of oocytes. Elevated expression of Spry2 mRNA in cumulus and mural granulosa cells correlated with human chorionic gonadotropin-induced expression of mRNAs encoding EGF-like peptides. Therefore, oocyte-derived members of the TGFbeta superfamily suppress FGF-stimulated Spry2 mRNA levels before the luteinizing hormone surge but promote Spry2 mRNA levels stimulated by EGF receptor-mediated signals after the surge.


Subject(s)
Cumulus Cells/metabolism , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Oocytes/metabolism , TGF-beta Superfamily Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Cumulus Cells/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental/drug effects , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Oocytes/drug effects , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TGF-beta Superfamily Proteins/pharmacology , Time Factors
13.
Dev Biol ; 317(1): 72-82, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18353305

ABSTRACT

A novel mutation, repro5, was isolated in a forward genetic screen for infertility mutations induced by ENU mutagenesis. Homozygous mutant mice were phenotypically normal but were infertile. Oocytes from mutant females appeared normal, but were severely maturation-defective in that they had reduced ability to progress to metaphase II (MII), and those reaching MII were unable to progress beyond the two pronuclei stage following in vitro fertilization (IVF). Mutant males exhibited defective spermiogenesis, resulting in oligoasthenoteratospermia. Genetic mapping, positional cloning, and complementation studies with a disruption allele led to the identification of a mutation in Brwd1 (Bromodomain and WD repeat domain containing 1) as the causative lesion. Bromodomain-containing proteins typically interact with regions of chromatin containing histones hyperacetylated at lysine residues, a characteristic of chromatin in early spermiogenesis before eventual replacement of histones by the protamines. Previous data indicated that Brwd1 is broadly expressed, encoding a putative transcriptional regulator that is believed to act on chromatin through interactions with the Brg1-dependent SWI/SNF chromatin-remodeling pathway. Brwd1 represents one of a small number of genes whose elimination disrupts gametogenesis in both sexes after the major events of meiotic prophase I have been completed.


Subject(s)
Oogenesis , Spermatogenesis , Animals , Cloning, Molecular , Ethylnitrosourea , Male , Meiosis , Mice , Mice, Inbred C57BL , Mutagenesis , Oocytes/cytology , Oocytes/metabolism , Sex Characteristics
14.
Development ; 135(1): 111-21, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18045843

ABSTRACT

Oocyte-derived bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) are key regulators of follicular development. Here we show that these factors control cumulus cell metabolism, particularly glycolysis and cholesterol biosynthesis before the preovulatory surge of luteinizing hormone. Transcripts encoding enzymes for cholesterol biosynthesis were downregulated in both Bmp15(-/-) and Bmp15(-/-) Gdf9(+/-) double mutant cumulus cells, and in wild-type cumulus cells after removal of oocytes from cumulus-cell-oocyte complexes. Similarly, cholesterol synthesized de novo was reduced in these cumulus cells. This indicates that oocytes regulate cumulus cell cholesterol biosynthesis by promoting the expression of relevant transcripts. Furthermore, in wild-type mice, Mvk, Pmvk, Fdps, Sqle, Cyp51, Sc4mol and Ebp, which encode enzymes required for cholesterol synthesis, were highly expressed in cumulus cells compared with oocytes; and oocytes, in the absence of the surrounding cumulus cells, synthesized barely detectable levels of cholesterol. Furthermore, coincident with reduced cholesterol synthesis in double mutant cumulus cells, lower levels were also detected in cumulus-cell-enclosed double mutant oocytes compared with wild-type oocytes. Levels of cholesterol synthesis in double mutant cumulus cells and oocytes were partially restored by co-culturing with wild-type oocytes. Together, these results indicate that mouse oocytes are deficient in synthesizing cholesterol and require cumulus cells to provide products of the cholesterol biosynthetic pathway. Therefore, oocyte-derived paracrine factors, particularly, BMP15 and GDF9, promote cholesterol biosynthesis in cumulus cells, probably as compensation for oocyte deficiencies in cholesterol production.


Subject(s)
Cholesterol/biosynthesis , Cumulus Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oocytes/metabolism , Animals , Bone Morphogenetic Protein 15 , Female , Gene Expression Regulation, Developmental , Growth Differentiation Factor 9 , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mutation/genetics , RNA, Messenger/genetics , Transcription, Genetic/genetics
15.
Development ; 134(14): 2593-603, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17553902

ABSTRACT

Mammalian oocytes are deficient in their ability to carry out glycolysis. Therefore, the products of glycolysis that are necessary for oocyte development are provided to oocytes by companion cumulus cells. Mouse oocytes secrete paracrine factors that promote glycolysis in cumulus cells. The objective of this study was to identify paracrine factors secreted by oocytes that promote glycolysis and expression of mRNA encoding the glycolytic enzymes PFKP and LDHA. Candidates included growth differentiation factor 9 (GDF9), bone morphogenetic protein 15 (BMP15) and fibroblast growth factors (FGFs). Bmp15-/- and Gdf9+/- Bmp15-/- (double mutant, DM) cumulus cells exhibited reduced levels of both glycolysis and Pfkp and Ldha mRNA, and mutant oocytes were deficient in promoting glycolysis and expression of Pfkp and Ldha mRNA in cumulus cells of wild-type (WT) mice. Alone, neither recombinant BMP15, GDF9 nor FGF8 promoted glycolysis and expression of Pfkp and Ldha mRNA in WT cumulus cells. Co-treatment with BMP15 and FGF8 promoted glycolysis and increased expression of Pfkp and Ldha mRNA in WT cumulus cells to the same levels as WT oocytes; however, the combinations of BMP15/GDF9 or GDF9/FGF8 did not. Furthermore, SU5402, an FGF receptor-dependent protein kinase inhibitor, inhibited Pfkp and Ldha expression in cumulus cells promoted by paracrine oocyte factors. Therefore, oocyte-derived BMP15 and FGFs cooperate to promote glycolysis in cumulus cells.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Granulosa Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oocytes/metabolism , Phosphofructokinase-1, Type C/metabolism , Animals , Bone Morphogenetic Protein 15 , Cells, Cultured , Female , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/pharmacology , Glycolysis , Granulosa Cells/drug effects , Growth Differentiation Factor 9 , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Mice , Mice, Mutant Strains , Oocytes/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology
16.
J Cell Sci ; 120(Pt 8): 1330-40, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17389684

ABSTRACT

The two principal functions of ovarian follicles are developmental and endocrine. The cumulus cells surrounding the oocyte are specialized to serve the development of the oocyte and steroidogenesis is a principal role of mural granulosa cells that line the follicle wall. The findings in this report demonstrate that oocytectomy or treatment with an inhibitor of SMAD2/3 activation results in decreased cumulus marker mRNA transcript levels and allows FSH to induce mural marker transcripts in cumulus cells. In addition, SMAD2/3 signaling is involved in enabling cumulus expansion and EGF-induced increases in Ptx3, Ptgs2 and Has2 mRNA levels. By contrast, follicle-stimulating hormone (FSH) stimulated expression of mural transcripts, but suppressed levels of cumulus transcripts. Thus, FSH and oocyte-stimulated SMAD2/3 signaling establish opposing gradients of influence in the follicle. These specify the mural and cumulus granulosa cell phenotypes that are pivotal for appropriate endocrine function and oocyte development.


Subject(s)
Cell Lineage , Oocytes/cytology , Ovarian Follicle/cytology , Animals , Benzamides/pharmacology , Cells, Cultured , Dioxoles/pharmacology , Female , Fluorescent Antibody Technique , MAP Kinase Signaling System , Mice , Ovarian Follicle/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/metabolism
17.
Dev Biol ; 302(1): 104-17, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17022963

ABSTRACT

There is massive destruction of transcripts during the maturation of mouse oocytes. The objective of this project was to identify and characterize the transcripts that are degraded versus those that are stable during the transcriptionally silent germinal vesicle (GV)-stage to metaphase II (MII)-stage transition using a microarray approach. A system for oocyte transcript amplification using both internal and 3'-poly(A) priming was utilized to minimize the impact of complex variations in transcript polyadenylation prevalent during this transition. Transcripts were identified and quantified using the Affymetrix Mouse Genome 430 v2.0 GeneChip. The significantly changed and stable transcripts were analyzed using Ingenuity Pathways Analysis and GenMAPP/MAPPFinder to characterize the biological themes underlying global changes in oocyte transcripts during maturation. It was concluded that the destruction of transcripts during the GV to MII transition is a selective rather than promiscuous process in mouse oocytes. In general, transcripts involved in processes that are associated with meiotic arrest at the GV-stage and the progression of oocyte maturation, such as oxidative phosphorylation, energy production, and protein synthesis and metabolism, were dramatically degraded. In contrast, transcripts encoding participants in signaling pathways essential for maintaining the unique characteristics of the MII-arrested oocyte, such as those involved in protein kinase pathways, were the most prominent among the stable transcripts.


Subject(s)
Meiosis/genetics , Oocytes/physiology , RNA Stability , RNA, Messenger/metabolism , Animals , Female , Gene Expression Profiling , Metabolic Networks and Pathways , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Ovarian Follicle/cytology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
18.
Dev Biol ; 286(2): 493-506, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16168984

ABSTRACT

The molecular basis for acquisition of meiotic and developmental competence, the two main outcomes of oocyte development and essential for producing an egg capable of being fertilized and supporting development to term, is largely unknown. Using microarrays, we characterized global changes in gene expression in oocytes derived from primordial, primary, secondary, small antral, and large antral follicles and used Expression Analysis Systematic Explorer (EASE) to identify biological and molecular processes that accompany these transitions and likely underpin acquisition of meiotic and developmental competence. The greatest degree of change in gene expression occurs during the primordial to primary follicle transition. Of particular interest is that specific chromosomes display significant changes in their overall transcriptional activity and that in some cases these changes are largely confined to specific regions on these chromosomes. We also examined the transcript profile of oocytes that developed in vitro, as well as following eCG priming. Remarkably, the expression profiles only differed by 4% and 2% from oocytes that developed in vivo when compared to oocytes that developed in vitro from either primordial or secondary follicles, respectively. About 1% of the genes were commonly mis-expressed, and EASE analysis revealed there is an over-representation of genes involved in transcription. Developmental competence of oocytes obtained from eCG-primed mice was substantially improved when compared to oocytes obtained from unprimed mice, and this correlated with decreased expression of genes implicated in basal transcription.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gonadotropins/pharmacology , Oocytes/growth & development , Animals , Embryonic Development , Mice , Oligonucleotide Array Sequence Analysis , Oocytes/drug effects , Oocytes/metabolism , RNA, Messenger/analysis , Transcription, Genetic/genetics
19.
Biol Reprod ; 73(2): 351-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15843493

ABSTRACT

A search for genes expressed more highly in mouse cumulus cells than mural granulosa cells by subtraction hybridization yielded Slc38a3. SLC38A3 is a sodium-coupled neutral amino acid transporter having substrate preference for l-glutamate, l-histidine, and l-alanine. Detectable levels of Slc38a3 mRNA were found by in situ hybridization in granulosa cells of large preantral follicles, but levels were higher in all granulosa cells of small antral follicles; expression became limited to cumulus cells of large antral follicles. Expression of Slc38a3 mRNA in granulosa cells was promoted by fully grown oocytes from antral follicles but not by growing oocytes from preantral follicles. Fully grown oocytes were dependent on cumulus cells for uptake of l-alanine and l-histidine but not l-leucine. Fully grown but not growing oocytes secreted one or more paracrine factors that promoted cumulus cell uptake of all three amino acids but of l-alanine and l-histidine to a much greater extent than l-leucine. Uptake of l-leucine appeared dependent primarily on contact-mediated signals from fully grown oocytes. Fully grown oocytes also promoted elevated levels of Slc38a3 mRNA and l-alanine transport by preantral granulosa cells, but growing oocytes did not. Therefore, fully grown oocytes secrete one or more paracrine factors that promote cumulus cell uptake of amino acids that oocytes themselves transport poorly. These amino acids are likely transferred to oocytes via gap junctions. Thus, oocytes use paracrine signals to promote their own development via metabolic cooperativity with cumulus cells. The ability of oocytes to mediate this cooperativity is developmentally regulated and acquired only in later stages of oocyte development.


Subject(s)
Amino Acid Transport System A/metabolism , Granulosa Cells/metabolism , Oocytes/metabolism , Alanine/metabolism , Amino Acid Transport System A/genetics , Animals , Female , Gap Junctions/metabolism , Histidine/metabolism , In Situ Hybridization , Leucine/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Xenopus
20.
Dev Biol ; 272(1): 1-14, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15242786

ABSTRACT

ATRX is a centromeric heterochromatin binding protein belonging to the SNF2 family of helicase/ATPases with chromatin remodeling activity. Mutations in the human ATRX gene result in X-linked alpha-thalassaemia with mental retardation (ATRX) syndrome and correlate with changes in methylation of repetitive DNA sequences. We show here that ATRX also functions to regulate key stages of meiosis in mouse oocytes. At the germinal vesicle (GV) stage, ATRX was found associated with the perinucleolar heterochromatin rim in transcriptionally quiescent oocytes. Phosphorylation of ATRX during meiotic maturation is dependent upon calcium calmodulin kinase (CamKII) activity. Meiotic resumption also coincides with deacetylation of histone H4 at lysine 5 (H4K5 Ac) while ATRX and histone H3 methylated on lysine 9 (H3K9) remained bound to the centromeres and interstitial regions of condensing chromosomes, respectively. Inhibition of histone deacetylases (HDACs) with trichostatin A (TSA) disrupted ATRX binding to the centromeres of hyperacetylated chromosomes resulting in abnormal chromosome alignments at metaphase II (MII). Similarly, while selective ablation of ATRX by antibody microinjection and RNA interference (RNAi) had no effect on the progression of meiosis, it had severe consequences for the alignment of chromosomes on the metaphase II spindle. These results suggest that genome-wide epigenetic modifications such as global histone deacetylation are essential for the binding of ATRX to centromeric heterochromatin. Moreover, centromeric ATRX is required for correct chromosome alignment and organization of a bipolar meiotic metaphase II spindle.


Subject(s)
DNA Helicases/metabolism , Meiosis/physiology , Metaphase/genetics , Nuclear Proteins/metabolism , Oocytes/physiology , Spindle Apparatus/genetics , Animals , Antibodies, Monoclonal/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly , Chromosomes/genetics , Chromosomes/metabolism , DNA Helicases/genetics , DNA Helicases/immunology , Enzyme Inhibitors/pharmacology , Female , Heterochromatin/metabolism , Histone Deacetylase Inhibitors , Histones , Hydroxamic Acids/pharmacology , Lysine/metabolism , Mice , Mice, Inbred Strains , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism , X-linked Nuclear Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...