Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 110(1): 102-115, 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-37774352

ABSTRACT

In response to luteinizing hormone (LH), multiple proteins in rat and mouse granulosa cells are rapidly dephosphorylated, but the responsible phosphatases remain to be identified. Because the phosphorylation state of phosphatases can regulate their interaction with substrates, we searched for phosphatases that might function in LH signaling by using quantitative mass spectrometry. We identified all proteins in rat ovarian follicles whose phosphorylation state changed detectably in response to a 30-min exposure to LH, and within this list, identified protein phosphatases or phosphatase regulatory subunits that showed changes in phosphorylation. Phosphatases in the phosphoprotein phosphatase (PPP) family were of particular interest because of their requirement for dephosphorylating the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase in the granulosa cells, which triggers oocyte meiotic resumption. Among the PPP family regulatory subunits, PPP1R12A and PPP2R5D showed the largest increases in phosphorylation, with 4-10 fold increases in signal intensity on several sites. Although follicles from mice in which these phosphorylations were prevented by serine-to-alanine mutations in either Ppp1r12a or Ppp2r5d showed normal LH-induced NPR2 dephosphorylation, these regulatory subunits and others could act redundantly to dephosphorylate NPR2. Our identification of phosphatases and other proteins whose phosphorylation state is rapidly modified by LH provides clues about multiple signaling pathways in ovarian follicles.


Subject(s)
Guanylate Cyclase , Phosphoric Monoester Hydrolases , Animals , Female , Mice , Rats , Guanylate Cyclase/metabolism , Luteinizing Hormone/metabolism , Meiosis , Oocytes/metabolism , Ovarian Follicle/metabolism , Phosphoric Monoester Hydrolases/metabolism
2.
Endocrinology ; 164(11)2023 09 23.
Article in English | MEDLINE | ID: mdl-37768169

ABSTRACT

More adolescents are coming out as transgender each year and are put on puberty blockers to suppress natal puberty, which is then followed by cross-hormone treatment to achieve puberty of the desired gender. Studies to examine the effects of puberty suppression and virilizing therapy on future reproductive potential among transgender males are lacking. This study used a translational murine in vitro fertilization model to examine the effects of female puberty suppression with depot leuprolide acetate (LA), followed by virilizing therapy with testosterone cypionate (T), on embryologic and pregnancy outcomes. LA effectively inhibited puberty when mice were treated beginning at 3 weeks of age. LA treatment was associated with higher mouse weight but lower ovarian weight. LA-treated mice ovulated developmentally competent eggs in response to gonadotropin administration, albeit at a higher dose than controls. Ovaries from mice treated with LA and T produced oocytes that had morphologically normal meiotic spindles after in vitro maturation and responded to gonadotropin stimulation. Eggs from mice treated with LA and T were fertilizable and produced developmentally competent embryos that led to births of fertile pups. These results suggest that fertility may not be impaired after puberty suppression and cross-hormone therapy for transgender males.


Subject(s)
Leuprolide , Sexual Maturation , Male , Female , Mice , Animals , Leuprolide/pharmacology , Leuprolide/therapeutic use , Testosterone/pharmacology , Gonadotropins , Ovary , Gonadotropin-Releasing Hormone
3.
bioRxiv ; 2023 Sep 16.
Article in English | MEDLINE | ID: mdl-37333193

ABSTRACT

In response to luteinizing hormone, multiple proteins in rat and mouse granulosa cells are rapidly dephosphorylated, but the responsible phosphatases remain to be identified. Because the phosphorylation state of phosphatases can regulate their interaction with substrates, we searched for phosphatases that might function in LH signaling by using quantitative mass spectrometry. We identified all proteins in rat ovarian follicles whose phosphorylation state changed detectably in response to a 30-minute exposure to LH, and within this list, identified protein phosphatases or phosphatase regulatory subunits that showed changes in phosphorylation. Phosphatases in the PPP family were of particular interest because of their requirement for dephosphorylating the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase in the granulosa cells, which triggers oocyte meiotic resumption. Among the PPP family regulatory subunits, PPP1R12A and PPP2R5D showed the largest increases in phosphorylation, with 4-10 fold increases in signal intensity on several sites. Although follicles from mice in which these phosphorylations were prevented by serine-to-alanine mutations in either Ppp1r12a or Ppp2r5d showed normal LH-induced NPR2 dephosphorylation, these regulatory subunits and others could act redundantly to dephosphorylate NPR2. Our identification of phosphatases and other proteins whose phosphorylation state is rapidly modified by LH provides clues about multiple signaling pathways in ovarian follicles. Summary sentence: Quantitative mass spectrometric analysis of phosphatases whose phosphorylation state is rapidly modified by luteinizing hormone provides clues about how LH signaling dephosphorylates NPR2 as well as a resource for future studies.

4.
J Assist Reprod Genet ; 40(5): 1215-1223, 2023 May.
Article in English | MEDLINE | ID: mdl-37058262

ABSTRACT

PURPOSE: This study was designed to determine if DMO limits in vitro development of aneuploid-enriched mouse embryos by activating a Trp53-dependent mechanism. METHODS: Mouse cleavage-stage embryos were treated with reversine to induce aneuploidy or vehicle to generate controls, and then cultured in media supplemented with DMO to reduce the pH of the culture media. Embryo morphology was assessed by phase microscopy. Cell number, mitotic figures, and apoptotic bodies were revealed by staining fixed embryos with DAPI. mRNA levels of Trp53, Oct-4, and Cdx2 were monitored by quantitative polymerase chain reactions (qPCRs). The effect of Trp53 on the expression of Oct-4 and Cdx2 was assessed by depleting Trp53 using Trp53 siRNA. RESULTS: Aneuploid-enriched late-stage blastocysts were morphologically indistinguishable from control blastocysts but had fewer cells and reduced mRNA levels of Oct-4 and Cdx2. Adding 1 mM DMO to the culture media during the 8-cell to blastocyst transition reduced the formation of aneuploid-enriched late-stage blastocysts but not control blastocysts and further suppressed the levels of Oct-4 and Cdx2 mRNA. Trp53 RNA levels in aneuploid-enriched embryos that were exposed to DMO were > twofold higher than controls, and Trp53 siRNA levels reduced the levels of Trp53 and increased levels of Oct-4 and Cdx2 mRNA by > twofold. CONCLUSION: These studies suggest that the development of morphologically normal aneuploid-enriched mouse blastocysts can be inhibited by adding low amounts of DMO to the culture media, which results in elevated levels of Trp53 mRNA that suppresses Oct-4 and Cdx2 expression.


Subject(s)
Blastocyst , Dimethadione , Mice , Animals , Dimethadione/metabolism , Blastocyst/metabolism , Aneuploidy , RNA, Messenger/metabolism , Culture Media/pharmacology , Culture Media/metabolism , Embryonic Development/genetics
5.
Front Mol Neurosci ; 15: 1007026, 2022.
Article in English | MEDLINE | ID: mdl-36340689

ABSTRACT

The natriuretic peptide receptors NPR1 and NPR2, also known as guanylyl cyclase A and guanylyl cyclase B, have critical functions in many signaling pathways, but much remains unknown about their localization and function in vivo. To facilitate studies of these proteins, we developed genetically modified mouse lines in which endogenous NPR1 and NPR2 were tagged with the HA epitope. To investigate the role of phosphorylation in regulating NPR1 and NPR2 guanylyl cyclase activity, we developed mouse lines in which regulatory serines and threonines were substituted with glutamates, to mimic the negative charge of the phosphorylated forms (NPR1-8E and NPR2-7E). Here we describe the generation and applications of these mice. We show that the HA-NPR1 and HA-NPR2 mice can be used to characterize the relative expression levels of these proteins in different tissues. We describe studies using the NPR2-7E mice that indicate that dephosphorylation of NPR2 transduces signaling pathways in ovary and bone, and studies using the NPR1-8E mice that indicate that the phosphorylation state of NPR1 is a regulator of heart, testis, and adrenal function.

6.
Endocrinology ; 161(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32384146

ABSTRACT

Meiotic arrest and resumption in mammalian oocytes are regulated by 2 opposing signaling proteins in the cells of the surrounding follicle: the guanylyl cyclase natriuretic peptide receptor 2 (NPR2), and the luteinizing hormone receptor (LHR). NPR2 maintains a meiosis-inhibitory level of cyclic guanosine 5'-monophosphate (cGMP) until LHR signaling causes dephosphorylation of NPR2, reducing NPR2 activity, lowering cGMP to a level that releases meiotic arrest. However, the signaling pathway between LHR activation and NPR2 dephosphorylation remains incompletely understood, due in part to imprecise information about the cellular localization of these 2 proteins. To investigate their localization, we generated mouse lines in which hemagglutinin epitope tags were added to the endogenous LHR and NPR2 proteins, and used immunofluorescence and immunogold microscopy to localize these proteins with high resolution. The results showed that the LHR protein is absent from the cumulus cells and inner mural granulosa cells, and is present in only 13% to 48% of the outer mural granulosa cells. In contrast, NPR2 is present throughout the follicle, and is more concentrated in the cumulus cells. Less than 20% of the NPR2 is in the same cells that express the LHR. These results suggest that to account for the LH-induced inactivation of NPR2, LHR-expressing cells send a signal that inactivates NPR2 in neighboring cells that do not express the LHR. An inhibitor of gap junction permeability attenuates the LH-induced cGMP decrease in the outer mural granulosa cells, consistent with this mechanism contributing to how NPR2 is inactivated in cells that do not express the LHR.


Subject(s)
Cyclic GMP/metabolism , Ovarian Follicle/enzymology , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, LH/metabolism , Animals , Female , Mice , Microscopy, Electron, Scanning , Ovarian Follicle/ultrastructure
7.
Biol Reprod ; 101(2): 338-346, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31201423

ABSTRACT

Mammalian oocytes are stored in the ovary for prolonged periods, and arrested in meiotic prophase. During this period, their plasma membranes are constantly being recycled by endocytosis and exocytosis. However, the function of this membrane turnover is unknown. Here, we investigated the requirement for exocytosis in the maintenance of meiotic arrest. Using Trim-away, a newly developed method for rapidly and specifically depleting proteins in oocytes, we have identified the SNARE protein, SNAP23, to be required for meiotic arrest. Degradation of SNAP23 causes premature meiotic resumption in follicle-enclosed oocytes. The reduction in SNAP23 is associated with loss of gap junction communication between the oocyte and surrounding follicle cells. Reduction of SNAP23 protein also inhibits regulated exocytosis in response to a Ca2+ stimulus (cortical granule exocytosis), as measured by lectin staining and cleavage of ZP2. Our results show an essential role for SNAP23 in two key processes that occur in mouse oocytes and eggs.


Subject(s)
Exocytosis/physiology , Oocytes/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Animals , Calcium/metabolism , Cell Culture Techniques , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Meiosis/physiology , Mice , Oocytes/drug effects , Ovarian Follicle , Preservatives, Pharmaceutical , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Thimerosal/pharmacology
8.
Biol Reprod ; 96(2): 341-351, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28203794

ABSTRACT

Embryonic poly(A)-binding protein (EPAB)-deficient mice are infertile due to defects in both the oocyte and the somatic cells of the ovary. Since EPAB is oocyte specific, the abnormalities in the somatic compartment of Epab−/− mice are likely due to factors inherent to the oocyte. Herein, we investigated whether oocyte­somatic communication is disrupted as a result of EPAB deficiency. We found that gap junctions are disrupted at the late preantral stage of folliculogenesis in Epab−/­ mice and remain disrupted in cumulus-enclosed oocytes (COCs) from antral follicles. Consistent with the timing of gap junction dysfunction, F-actin staining of transzonal processes (TZPs) is lower in Epab−/− follicles at the late preantral stage and completely absent in Epab−/− COCs. Epab−/− oocytes express significantly lower levels of the junction protein E-cadherin, which is likely to be a contributing factor leading to premature TZP retraction. Overall, these results demonstrate that EPAB is important for oocyte­somatic communication by maintaining TZPs and gap junctions at the preantral stage of folliculogenesis.


Subject(s)
Gene Expression Regulation/physiology , Oocytes/physiology , Ovarian Follicle/metabolism , Poly(A)-Binding Proteins/metabolism , Animals , Cell Communication/physiology , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Female , Gap Junctions/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Proteins/genetics , Gap Junction alpha-4 Protein
9.
Endocrinology ; 157(1): 405-16, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26492470

ABSTRACT

Embryonic poly(A)-binding protein (EPAB) is the predominant poly(A)-binding protein in Xenopus, mouse, and human oocytes and early embryos before zygotic genome activation. EPAB is required for translational activation of maternally stored mRNAs in the oocyte and Epab(-/-) female mice are infertile due to impaired oocyte maturation, cumulus expansion, and ovulation. The aim of this study was to characterize the mechanism of follicular somatic cell dysfunction in Epab(-/-) mice. Using a coculture system of oocytectomized cumulus oophorus complexes (OOXs) with denuded oocytes, we found that when wild-type OOXs were cocultured with Epab(-/-) oocytes, or when Epab(-/-) OOXs were cocultured with WT oocytes, cumulus expansion failed to occur in response to epidermal growth factor (EGF). This finding suggests that oocytes and cumulus cells (CCs) from Epab(-/-) mice fail to send and receive the necessary signals required for cumulus expansion. The abnormalities in Epab(-/-) CCs are not due to lower expression of the oocyte-derived factors growth differentiation factor 9 or bone morphogenetic protein 15, because Epab(-/-) oocytes express these proteins at comparable levels with WT. Epab(-/-) granulosa cells (GCs) exhibit decreased levels of phosphorylated MEK1/2, ERK1/2, and p90 ribosomal S6 kinase in response to lutenizing hormone and EGF treatment, as well as decreased phosphorylation of the EGF receptor. In conclusion, EPAB, which is oocyte specific, is required for the ability of CCs and GCs to become responsive to LH and EGF signaling. These results emphasize the importance of oocyte-somatic communication for GC and CC function.


Subject(s)
Cumulus Cells/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/agonists , Granulosa Cells/metabolism , Oocytes/metabolism , Poly(A)-Binding Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Protein 15/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cumulus Cells/cytology , ErbB Receptors/metabolism , Female , Granulosa Cells/cytology , Growth Differentiation Factor 9/metabolism , Luteinizing Hormone/metabolism , MAP Kinase Signaling System , Mice, Knockout , Oocytes/cytology , Phosphorylation , Poly(A)-Binding Proteins/genetics , Protein Processing, Post-Translational , Receptors, LH/agonists , Receptors, LH/metabolism
10.
Biol Reprod ; 93(2): 43, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26134869

ABSTRACT

During oocyte maturation, fertilization, and early embryo development until zygotic genome activation (ZGA), transcription is suppressed, and gene expression is dependent upon the timely activation of stored mRNAs. Embryonic poly(A)-binding protein (EPAB) is the predominant poly(A)-binding protein in Xenopus, mouse, and human oocytes and early embryos and is important for regulating translational activation of maternally stored mRNAs. EPAB is critical for early development because Epab(-/-) female mice do not produce mature eggs and are infertile. In this study, we further characterize morphological and molecular aspects of Epab(-/-) oocytes. We demonstrated that Epab(-/-) oocytes are smaller in size, contain peripheral germinal vesicles, and are loosely associated with cumulus cells. The chromatin reorganization of the surrounded nucleolus (SN) configuration and transcriptional silencing that normally occurs during oocyte growth does not occur in Epab(-/-) oocytes. Interestingly, microinjection of Epab mRNA into Epab(-/-) preantral follicle-enclosed oocytes rescues reorganization of chromatin and oocyte maturation to metaphase II. Overall, these results demonstrate an important role for EPAB during oocyte growth and the acquisition of meiotic competence.


Subject(s)
Chromatin/physiology , Meiosis/genetics , Oocytes/growth & development , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/physiology , Animals , Cell Nucleolus/genetics , Cell Nucleolus/ultrastructure , Female , Gene Silencing , Metaphase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/ultrastructure , Ovarian Follicle/growth & development , Ovarian Follicle/ultrastructure , Pregnancy , Protein Modification, Translational/genetics , Spindle Apparatus/genetics
11.
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
13.
PLoS One ; 8(6): e65365, 2013.
Article in English | MEDLINE | ID: mdl-23826079

ABSTRACT

G protein-coupled receptor 3 (GPR3) is a constitutively active receptor that maintains high 3'-5'-cyclic adenosine monophosphate (cAMP) levels required for meiotic arrest in oocytes and CNS function. Ligand-activated G protein-coupled receptors (GPCRs) signal at the cell surface and are silenced by phosphorylation and ß-arrestin recruitment upon endocytosis. Some GPCRs can also signal from endosomes following internalization. Little is known about the localization, signaling, and regulation of constitutively active GPCRs. We demonstrate herein that exogenously-expressed GPR3 localizes to the cell membrane and undergoes internalization in HEK293 cells. Inhibition of endocytosis increased cell surface-localized GPR3 and cAMP levels while overexpression of GPCR-Kinase 2 (GRK2) and ß-arrestin-2 decreased cell surface-localized GPR3 and cAMP levels. GRK2 by itself is sufficient to decrease cAMP production but both GRK2 and ß-arrestin-2 are required to decrease cell surface GPR3. GRK2 regulates GPR3 independently of its kinase activity since a kinase inactive GRK2-K220R mutant significantly decreased cAMP levels. However, GRK2-K220R and ß-arrestin-2 do not diminish cell surface GPR3, suggesting that phosphorylation is required to induce GPR3 internalization. To understand which residues are targeted for desensitization, we mutated potential phosphorylation sites in the third intracellular loop and C-terminus and examined the effect on cAMP and receptor surface localization. Mutation of residues in the third intracellular loop dramatically increased cAMP levels whereas mutation of residues in the C-terminus produced cAMP levels comparable to GPR3 wild type. Interestingly, both mutations significantly reduced cell surface expression of GPR3. These results demonstrate that GPR3 signals at the plasma membrane and can be silenced by GRK2/ß-arrestin overexpression. These results also strongly implicate the serine and/or threonine residues in the third intracellular loop in the regulation of GPR3 activity.


Subject(s)
Cell Membrane/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 2/metabolism , Animals , Cell Membrane/drug effects , Cyclic AMP/metabolism , Dynamins/genetics , Dynamins/metabolism , Endocytosis/drug effects , Endocytosis/physiology , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Mutation , Phosphorylation , Protein Domains , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/genetics , Signal Transduction
14.
Biochem J ; 446(1): 47-58, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22621333

ABSTRACT

Gene expression during oocyte maturation and early embryogenesis up to zygotic genome activation requires translational activation of maternally-derived mRNAs. EPAB [embryonic poly(A)-binding protein] is the predominant poly(A)-binding protein during this period in Xenopus, mouse and human. In Xenopus oocytes, ePAB stabilizes maternal mRNAs and promotes their translation. To assess the role of EPAB in mammalian reproduction, we generated Epab-knockout mice. Although Epab(-/-) males and Epab(+/-) of both sexes were fertile, Epab(-/-) female mice were infertile, and could not generate embryos or mature oocytes in vivo or in vitro. Epab(-/-) oocytes failed to achieve translational activation of maternally-stored mRNAs upon stimulation of oocyte maturation, including Ccnb1 (cyclin B1) and Dazl (deleted in azoospermia-like) mRNAs. Microinjection of Epab mRNA into Epab(-/-) germinal vesicle stage oocytes did not rescue maturation, suggesting that EPAB is also required for earlier stages of oogenesis. In addition, late antral follicles in the ovaries of Epab(-/-) mice exhibited impaired cumulus expansion, and a 8-fold decrease in ovulation, associated with a significant down-regulation of mRNAs encoding the EGF (epidermal growth factor)-like growth factors Areg (amphiregulin), Ereg (epiregulin) and Btc (betacellulin), and their downstream regulators, Ptgs2 (prostaglandin synthase 2), Has2 (hyaluronan synthase 2) and Tnfaip6 (tumour necrosis factor α-induced protein 6). The findings from the present study indicate that EPAB is necessary for oogenesis, folliculogenesis and female fertility in mice.


Subject(s)
Fertility/physiology , Oocytes/physiology , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Animals , Base Sequence , Cyclin B1 , Female , Fertility/genetics , Gene Expression Regulation , Infertility, Female/genetics , Male , Metaphase/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Oogenesis/physiology , Ovarian Follicle/physiology , Ovulation/genetics , Poly(A)-Binding Protein I/genetics , Polyadenylation , Spindle Apparatus/genetics
15.
Matrix Biol ; 31(5): 299-307, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22525512

ABSTRACT

There is considerable interest in understanding prostate cancer metastasis to bone and the interaction of these cells with the bone microenvironment. Osteonectin/SPARC/BM-40 is a collagen binding matricellular protein that is enriched in bone. Its expression is increased in prostate cancer metastases, and it stimulates the migration of prostate carcinoma cells. However, the presence of osteonectin in cancer cells and the stroma may limit prostate tumor development and progression. To determine how bone matrix osteonectin affects the behavior of prostate cancer cells, we modeled prostate cancer cell-bone interactions using the human prostate cancer cell line PC-3, and mineralized matrices synthesized by wild type and osteonectin-null osteoblasts in vitro. We developed this in vitro system because the structural complexity of collagen matrices in vivo is not mimicked by reconstituted collagen scaffolds or by more complex substrates, like basement membrane extracts. Second harmonic generation imaging demonstrated that the wild type matrices had thick collagen fibers organized into longitudinal bundles, whereas osteonectin-null matrices had thinner fibers in random networks. Importantly, a mouse model of prostate cancer metastases to bone showed a collagen fiber phenotype similar to the wild type matrix synthesized in vitro. When PC-3 cells were grown on the wild type matrices, they displayed decreased cell proliferation, increased cell spreading, and decreased resistance to radiation-induced cell death, compared to cells grown on osteonectin-null matrix. Our data support the idea that osteonectin can suppress prostate cancer pathogenesis, expanding this concept to the microenvironment of skeletal metastases.


Subject(s)
Bone Matrix/metabolism , Gamma Rays , Osteoblasts/drug effects , Osteonectin/metabolism , Prostatic Neoplasms/pathology , Animals , Blotting, Western , Bone Matrix/drug effects , Bone Matrix/pathology , Bone Matrix/radiation effects , Cell Communication , Cell Death/radiation effects , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Shape , Cell Survival , Male , Mice , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoblasts/radiation effects , Osteonectin/genetics , Osteonectin/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tumor Microenvironment
16.
Reproduction ; 141(6): 737-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411693

ABSTRACT

Mammalian oocytes are arrested at prophase I of meiosis until a preovulatory surge of LH stimulates them to resume meiosis. Prior to the LH surge, high levels of cAMP within the oocyte maintain meiotic arrest; this cAMP is generated in the oocyte through the activity of the constitutively active, G(s)-coupled receptor, G-protein-coupled receptor 3 (GPR3) or GPR12. Activated GPRs are typically targeted for desensitization through receptor-mediated endocytosis, but a continuously high level of cAMP is needed for meiotic arrest. The aim of this study was to examine whether receptor-mediated endocytosis occurs in the mouse oocyte and whether this could affect the maintenance of meiotic arrest. We found that constitutive endocytosis occurs in the mouse oocyte. Inhibitors of receptor-mediated endocytosis, monodansylcadaverine and dynasore, inhibited the formation of early endosomes and completely inhibited spontaneous meiotic resumption. A red fluorescent protein-tagged GPR3 localized in the plasma membrane and within early endosomes in the oocyte, demonstrating that GPR3 is endocytosed. However, overexpression of G-protein receptor kinase 2 and ß-arrestin-2 had only a modest effect on stimulating meiotic resumption, suggesting that these proteins do not play a major role in GPR3 endocytosis. Inhibition of endocytosis elevated cAMP levels within oocytes, suggesting that there is an accumulation of GPR3 at the plasma membrane. These results show that endocytosis occurs in the oocyte, leading to a decrease in cAMP production, and suggest that there is a balance between cAMP production and degradation in the arrested oocyte that maintains cAMP levels at an appropriate level during the maintenance of meiotic arrest.


Subject(s)
Cell Membrane/metabolism , Cyclic AMP/metabolism , Endocytosis , Meiotic Prophase I , Oocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Second Messenger Systems , Analysis of Variance , Animals , Arrestins/genetics , Arrestins/metabolism , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Membrane/drug effects , Cells, Cultured , Endocytosis/drug effects , Endosomes/metabolism , Female , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Hydrazones/pharmacology , Meiotic Prophase I/drug effects , Mice , Oocytes/drug effects , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/drug effects , Time Factors , Transfection , beta-Arrestin 2 , beta-Arrestins
17.
Biol Reprod ; 83(4): 578-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20610804

ABSTRACT

Oocyte maturation in rodents is characterized by a dramatic reorganization of the endoplasmic reticulum (ER) and an increase in the ability of an oocyte to release Ca(2+) in response to fertilization or inositol 1,4,5-trisphosphate (IP(3)). We examined if human oocytes undergo similar changes during cytoplasmic meiotic maturation both in vivo and in vitro. Immature, germinal vesicle (GV)-stage oocytes had a fine network of ER throughout the cortex and interior, whereas the ER in the in vivo-matured, metaphase II oocytes was organized in large (diameter, ∼2-3 µm) accumulations throughout the cortex and interior. Likewise, oocytes matured in vitro exhibited cortical and interior clusters with no apparent polarity in regard to the meiotic spindle. In vivo-matured oocytes contained approximately 1.5-fold the amount of IP(3) receptor protein and released significantly more Ca(2+) in response to IP(3) compared with GV-stage oocytes; however, oocytes matured in vitro did not contain more IP(3) receptor protein or release more Ca(2+) in response to IP(3) compared with GV-stage oocytes. These results show that at least one cytoplasmic change occurs during in vitro maturation of human oocytes that might be important for fertilization and subsequent embryonic development, but they suggest that a low developmental competence of in vitro-matured oocytes could be the result of deficiencies in the ability to release Ca(2+) at fertilization.


Subject(s)
Calcium/physiology , Endoplasmic Reticulum/physiology , Meiosis/physiology , Oocytes/physiology , Adult , Animals , Blotting, Western , Cell Polarity/physiology , Endoplasmic Reticulum/ultrastructure , Female , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Mice , Microscopy, Confocal , Oocytes/ultrastructure , Young Adult
18.
Biol Reprod ; 81(1): 147-54, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19299317

ABSTRACT

Oocyte cryopreservation is a promising technology that could benefit women undergoing assisted reproduction. Most studies examining the effects of cryopreservation on fertilization and developmental competence have been done using metaphase II-stage oocytes, while fewer studies have focused on freezing oocytes at the germinal vesicle (GV) stage, followed by in vitro maturation. Herein, we examined the effects of vitrifying GV-stage mouse oocytes on cytoplasmic structure and on the ability to undergo cytoplasmic changes necessary for proper fertilization and early embryonic development. We examined the endoplasmic reticulum (ER) as one indicator of cytoplasmic structure, as well as the ability of oocytes to develop Ca(2+) release mechanisms following vitrification and in vitro maturation. Vitrified GV-stage oocytes matured in culture to metaphase II at a rate comparable to that of controls. These oocytes had the capacity to release Ca(2+) following injection of inositol 1,4,5-trisphosphate, demonstrating that Ca(2+) release mechanisms developed during meiotic maturation. The ER remained intact during the vitrification procedure as assessed using the lipophilic fluorescent dye DiI. However, the reorganization of the ER that occurs during in vivo maturation was impaired in oocytes that were vitrified before oocyte maturation. These results show that vitrification of GV-stage oocytes does not affect nuclear maturation or the continuity of the ER, but normal cytoplasmic maturation as assessed by the reorganization of the ER is disrupted. Deficiencies in factors that are responsible for proper ER reorganization during oocyte maturation could contribute to the low developmental potential previously reported in vitrified in vitro-matured oocytes.


Subject(s)
Cryopreservation , Endoplasmic Reticulum/physiology , Fertilization/physiology , Oocytes , Oogenesis/physiology , Animals , Cell Culture Techniques , Cells, Cultured , Female , Fertilization in Vitro , Freezing/adverse effects , Meiosis/physiology , Mice , Oocytes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...