Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Dev Biol ; 434(1): 196-205, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29274320

RESUMO

Mammalian oocytes are arrested in meiotic prophase from around the time of birth until just before ovulation. Following an extended period of growth, they are stimulated to mature to the metaphase II stage by a preovulatory luteinizing hormone (LH) surge that occurs with each reproductive cycle. Small, growing oocytes are not competent to mature into fertilizable eggs because they do not possess adequate amounts of cell cycle regulatory proteins, particularly cyclin-dependent kinase 1 (CDK1). As oocytes grow, they synthesize CDK1 and acquire the ability to mature. After oocytes achieve meiotic competence, meiotic arrest at the prophase stage is dependent on high levels of cAMP that are generated in the oocyte under the control of the constitutively active Gs-coupled receptor, GPR3. In this study, we examined the switch between GPR3-independent and GPR3-dependent meiotic arrest. We found that the ability of oocytes to mature, as well as oocyte CDK1 levels, were dependent on follicle size, but CDK1 expression in oocytes from preantral follicles was not acutely altered by the activity of follicle stimulating hormone (FSH). Gpr3 was expressed and active in incompetent oocytes within early stage follicles, well before cAMP is required to maintain meiotic arrest. Oocytes from Gpr3-/- mice were less competent to mature than oocytes from Gpr3+/+ mice, as assessed by the time course of germinal vesicle breakdown. Correspondingly, Gpr3-/- oocytes contained significantly lower CDK1 levels than their Gpr3+/+ counterparts that were at the same stage of follicle development. These results demonstrate that GPR3 potentiates meiotic competence, most likely by raising cAMP.


Assuntos
Proteína Quinase CDC2/biossíntese , Pontos de Checagem do Ciclo Celular/fisiologia , AMP Cíclico/metabolismo , Regulação da Expressão Gênica/fisiologia , Prófase Meiótica I/fisiologia , Oócitos/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Sistemas do Segundo Mensageiro/fisiologia , Animais , Proteína Quinase CDC2/genética , AMP Cíclico/genética , Feminino , Camundongos , Camundongos Knockout , Oócitos/citologia , Receptores Acoplados a Proteínas G/genética
2.
Biol Reprod ; 101(2): 338-346, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31201423

RESUMO

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.


Assuntos
Exocitose/fisiologia , Oócitos/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Animais , Cálcio/metabolismo , Técnicas de Cultura de Células , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Meiose/fisiologia , Camundongos , Oócitos/efeitos dos fármacos , Folículo Ovariano , Conservantes Farmacêuticos , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Timerosal/farmacologia
3.
Development ; 142(15): 2633-40, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26160904

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Óvulo/fisiologia , Proteínas RGS/metabolismo , Interações Espermatozoide-Óvulo/fisiologia , Animais , Feminino , Imunofluorescência , Immunoblotting , Camundongos , Óvulo/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas
4.
Biol Reprod ; 93(2): 43, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26134869

RESUMO

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.


Assuntos
Cromatina/fisiologia , Meiose/genética , Oócitos/crescimento & desenvolvimento , Proteínas de Ligação a Poli(A)/genética , Proteínas de Ligação a Poli(A)/fisiologia , Animais , Nucléolo Celular/genética , Nucléolo Celular/ultraestrutura , Feminino , Inativação Gênica , Metáfase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/ultraestrutura , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/ultraestrutura , Gravidez , Modificação Traducional de Proteínas/genética , Fuso Acromático/genética
5.
Biochem J ; 446(1): 47-58, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22621333

RESUMO

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.


Assuntos
Fertilidade/fisiologia , Oócitos/fisiologia , Proteínas de Ligação a Poli(A)/genética , Proteínas de Ligação a Poli(A)/metabolismo , Animais , Sequência de Bases , Ciclina B1 , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Infertilidade Feminina/genética , Masculino , Metáfase/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Oogênese/fisiologia , Folículo Ovariano/fisiologia , Ovulação/genética , Proteína I de Ligação a Poli(A)/genética , Poliadenilação , Fuso Acromático/genética
6.
Endocrinology ; 164(11)2023 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-37768169

RESUMO

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.


Assuntos
Leuprolida , Maturidade Sexual , Masculino , Feminino , Camundongos , Animais , Leuprolida/farmacologia , Leuprolida/uso terapêutico , Testosterona/farmacologia , Gonadotropinas , Ovário , Hormônio Liberador de Gonadotropina
7.
Reproduction ; 141(6): 737-47, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21411693

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Endocitose , Prófase Meiótica I , Oócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistemas do Segundo Mensageiro , Análise de Variância , Animais , Arrestinas/genética , Arrestinas/metabolismo , Cadaverina/análogos & derivados , Cadaverina/farmacologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Endocitose/efeitos dos fármacos , Endossomos/metabolismo , Feminino , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Hidrazonas/farmacologia , Prófase Meiótica I/efeitos dos fármacos , Camundongos , Oócitos/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Fatores de Tempo , Transfecção , beta-Arrestina 2 , beta-Arrestinas
8.
Biol Reprod ; 83(4): 578-83, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20610804

RESUMO

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.


Assuntos
Cálcio/fisiologia , Retículo Endoplasmático/fisiologia , Meiose/fisiologia , Oócitos/fisiologia , Adulto , Animais , Western Blotting , Polaridade Celular/fisiologia , Retículo Endoplasmático/ultraestrutura , Feminino , Humanos , Inositol 1,4,5-Trifosfato/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Camundongos , Microscopia Confocal , Oócitos/ultraestrutura , Adulto Jovem
9.
J Cell Biol ; 171(2): 255-65, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16247026

RESUMO

The arrest of meiotic prophase in mouse oocytes within antral follicles requires the G protein G(s) and an orphan member of the G protein-coupled receptor family, GPR3. To determine whether GPR3 activates G(s), the localization of Galpha(s) in follicle-enclosed oocytes from Gpr3(+/+) and Gpr3(-/-) mice was compared by using immunofluorescence and Galpha(s)GFP. GPR3 decreased the ratio of Galpha(s) in the oocyte plasma membrane versus the cytoplasm and also decreased the amount of Galpha(s) in the oocyte. Both of these properties indicate that GPR3 activates G(s). The follicle cells around the oocyte are also necessary to keep the oocyte in prophase, suggesting that they might activate GPR3. However, GPR3-dependent G(s) activity was similar in follicle-enclosed and follicle-free oocytes. Thus, the maintenance of prophase arrest depends on the constitutive activity of GPR3 in the oocyte, and the follicle cell signal acts by a means other than increasing GPR3 activity.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Meiose/fisiologia , Oócitos/metabolismo , Prófase/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Oócitos/citologia , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia
10.
iScience ; 23(9): 101523, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32927266

RESUMO

HUWE1 is a HECT-domain ubiquitin E3 ligase expressed in various tissues. Although HUWE1 is known to promote degradation of the tumor suppressor p53, given a growing list of its substrates, in vivo functions of HUWE1 remain elusive. Here, we investigated the role of HUWE1 in the female reproductive system. Homozygous deletion of Huwe1 in mouse oocytes of primary follicles caused oocyte death and female infertility, whereas acute depletion of HUWE1 protein by Trim-Away technology did not impact oocytes from antral follicles. Interestingly, oocytes from Huwe1 heterozygous females matured and fertilized normally, but the majority of embryos that lacked maternal Huwe1 were arrested at the morula stage after fertilization. Consequently, Huwe1 heterozygous females only produced wild-type pups. Concomitant knockout of p53 did not recover fertility of the Huwe1 knockout females. These findings make HUWE1 a unique and critical maternal factor indispensable for maintaining the quality of oocytes and embryos.

11.
Dev Biol ; 316(1): 124-34, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18280465

RESUMO

Although it is established that cAMP accumulation plays a pivotal role in preventing meiotic resumption in mammalian oocytes, the mechanisms controlling cAMP levels in the female gamete have remained elusive. Both production of cAMP via GPCRs/Gs/adenylyl cyclases endogenous to the oocyte as well as diffusion from the somatic compartment through gap junctions have been implicated in maintaining cAMP at levels that preclude maturation. Here we have used a genetic approach to investigate the different biochemical pathways contributing to cAMP accumulation and maturation in mouse oocytes. Because cAMP hydrolysis is greatly decreased and cAMP accumulates above a threshold, oocytes deficient in PDE3A do not resume meiosis in vitro or in vivo, resulting in complete female infertility. In vitro, inactivation of Gs or downregulation of the GPCR GPR3 causes meiotic resumption in the Pde3a null oocytes. Crossing of Pde3a(-/-) mice with Gpr3(-/-) mice causes partial recovery of female fertility. Unlike the complete meiotic block of the Pde3a null mice, oocyte maturation is restored in the double knockout, although it occurs prematurely as described for the Gpr3(-/-) mouse. The increase in cAMP that follows PDE3A ablation is not detected in double mutant oocytes, confirming that GPR3 functions upstream of PDE3A in the regulation of oocyte cAMP. Metabolic coupling between oocytes and granulosa cells was not affected in follicles from the single or double mutant mice, suggesting that diffusion of cAMP is not prevented. Finally, simultaneous ablation of GPR12, an additional receptor expressed in the oocyte, does not modify the Gpr3(-/-) phenotype. Taken together, these findings demonstrate that Gpr3 is epistatic to Pde3a and that fertility as well as meiotic arrest in the PDE3A-deficient oocyte is dependent on the activity of GPR3. These findings also suggest that cAMP diffusion through gap junctions or the activity of additional receptors is not sufficient by itself to maintain the meiotic arrest in the mouse oocyte.


Assuntos
AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Meiose , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , AMP Cíclico/deficiência , AMP Cíclico/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Feminino , Fertilidade/genética , Junções Comunicantes/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Meiose/genética , Camundongos , Camundongos Mutantes , Oócitos/citologia , Ovário/citologia , Ovário/fisiologia , Receptores Acoplados a Proteínas G/genética , Xenopus laevis
12.
Methods Mol Biol ; 518: 157-73, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19085139

RESUMO

The mammalian oocyte develops within a complex of somatic cells known as a follicle, within which signals from the somatic cells regulate the oocyte, and signals from the oocyte regulate the somatic cells. Because isolation of the oocyte from the follicle disrupts these communication pathways, oocyte physiology is best studied within an intact follicle. Here we describe methods for quantitative microinjection of follicle-enclosed mouse oocytes, thus allowing the introduction of signaling molecules as well as optical probes into the oocyte within its physiological environment.


Assuntos
Microinjeções/métodos , Oócitos/metabolismo , Folículo Ovariano/citologia , Animais , Dióxido de Carbono , Células Cultivadas , Dissecação , Feminino , Umidade , Camundongos , Oócitos/citologia , Folículo Ovariano/metabolismo , Temperatura
14.
PLoS One ; 8(6): e65365, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826079

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 2/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Células HEK293 , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Mutação , Fosforilação , Domínios Proteicos , Inibidores de Proteínas Quinases/farmacologia , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais
15.
Biol Reprod ; 81(1): 147-54, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19299317

RESUMO

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.


Assuntos
Criopreservação , Retículo Endoplasmático/fisiologia , Fertilização/fisiologia , Oócitos , Oogênese/fisiologia , Animais , Técnicas de Cultura de Células , Células Cultivadas , Feminino , Fertilização in vitro , Congelamento/efeitos adversos , Meiose/fisiologia , Camundongos , Oócitos/ultraestrutura
16.
Development ; 136(11): 1869-78, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429786

RESUMO

Mammalian oocytes are arrested in meiotic prophase by an inhibitory signal from the surrounding somatic cells in the ovarian follicle. In response to luteinizing hormone (LH), which binds to receptors on the somatic cells, the oocyte proceeds to second metaphase, where it can be fertilized. Here we investigate how the somatic cells regulate the prophase-to-metaphase transition in the oocyte, and show that the inhibitory signal from the somatic cells is cGMP. Using FRET-based cyclic nucleotide sensors in follicle-enclosed mouse oocytes, we find that cGMP passes through gap junctions into the oocyte, where it inhibits the hydrolysis of cAMP by the phosphodiesterase PDE3A. This inhibition maintains a high concentration of cAMP and thus blocks meiotic progression. LH reverses the inhibitory signal by lowering cGMP levels in the somatic cells (from approximately 2 microM to approximately 80 nM at 1 hour after LH stimulation) and by closing gap junctions between the somatic cells. The resulting decrease in oocyte cGMP (from approximately 1 microM to approximately 40 nM) relieves the inhibition of PDE3A, increasing its activity by approximately 5-fold. This causes a decrease in oocyte cAMP (from approximately 700 nM to approximately 140 nM), leading to the resumption of meiosis.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/fisiologia , Meiose/fisiologia , Oócitos/fisiologia , Animais , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Feminino , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Humanos , Hormônio Luteinizante/farmacologia , Hormônio Luteinizante/fisiologia , Meiose/efeitos dos fármacos , Camundongos , Oócitos/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia
17.
Biol Reprod ; 78(4): 667-72, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18184921

RESUMO

In mammalian oocytes, the maintenance of meiotic prophase I arrest prior to the surge of LH that stimulates meiotic maturation depends on a high level of cAMP within the oocyte. In mouse and rat, the cAMP is generated in the oocyte, and this requires the activity of a constitutively active, Gs-linked receptor, GPR3 or GPR12, respectively. To examine if human oocyte meiotic arrest depends on a similar pathway, we used RT-PCR and Western blotting to look at whether human oocytes express the same components for maintaining arrest as rodent oocytes. RNA encoding GPR3, but not GPR12, was expressed. RNA encoding adenylate cyclase type 3, which is the major adenylate cyclase required for maintaining meiotic arrest in the mouse oocyte, was also expressed, as was Galphas protein. To determine if this pathway is functional in the human oocyte, we examined the effect of injecting a function-blocking antibody against Galphas on meiotic resumption. This antibody stimulated meiotic resumption of human oocytes that were maintained at the prophase I stage using a phosphodiesterase inhibitor. These results demonstrate that human oocytes maintain meiotic arrest prior to the LH surge using a signaling pathway similar to that of rodent oocytes.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Prófase Meiótica I/fisiologia , Oócitos/citologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/genética , Animais , Anticorpos/farmacologia , Western Blotting , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/imunologia , Humanos , Hormônio Luteinizante/metabolismo , Camundongos , Oócitos/química , RNA/análise , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Development ; 135(19): 3229-38, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18776144

RESUMO

Luteinizing hormone (LH) acts on ovarian follicles to reinitiate meiosis in prophase-arrested mammalian oocytes, and this has been proposed to occur by interruption of a meioisis-inhibitory signal that is transmitted through gap junctions into the oocyte from the somatic cells that surround it. To investigate this idea, we microinjected fluorescent tracers into live antral follicle-enclosed mouse oocytes, and we demonstrate for the first time that LH causes a decrease in the gap junction permeability between the somatic cells, prior to nuclear envelope breakdown (NEBD). The decreased permeability results from the MAP kinase-dependent phosphorylation of connexin 43 on serines 255, 262 and 279/282. We then tested whether the inhibition of gap junction communication was sufficient and necessary for the reinitiation of meiosis. Inhibitors that reduced gap junction permeability caused NEBD, but an inhibitor of MAP kinase activation that blocked gap junction closure in response to LH did not prevent NEBD. Thus, both MAP kinase-dependent gap junction closure and another redundant pathway function in parallel to ensure that meiosis resumes in response to LH.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Meiose/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Conexina 43/química , Células do Cúmulo/citologia , Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/metabolismo , Feminino , Junções Comunicantes/metabolismo , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Meiose/fisiologia , Camundongos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/citologia , Fosforilação , Serina/química , Técnicas de Cultura de Tecidos
19.
Dev Biol ; 310(2): 240-9, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17850783

RESUMO

The maintenance of meiotic prophase arrest in fully grown vertebrate oocytes depends on the activity of a G(s) G-protein that activates adenylyl cyclase and elevates cAMP, and in the mouse oocyte, G(s) is activated by a constitutively active orphan receptor, GPR3. To determine whether the action of luteinizing hormone (LH) on the mouse ovarian follicle causes meiotic resumption by inhibiting GPR3-G(s) signaling, we examined the effect of LH on the localization of Galpha(s). G(s) activation in response to stimulation of an exogenously expressed beta(2)-adrenergic receptor causes Galpha(s) to move from the oocyte plasma membrane into the cytoplasm, whereas G(s) inactivation in response to inhibition of the beta(2)-adrenergic receptor causes Galpha(s) to move back to the plasma membrane. However, LH does not cause a change in Galpha(s) localization, indicating that LH does not act by terminating receptor-G(s) signaling.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Hormônio Luteinizante/farmacologia , Meiose/fisiologia , Oócitos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Cruzamentos Genéticos , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , Transporte Proteico , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais
20.
Dev Biol ; 299(2): 345-55, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16949564

RESUMO

The signaling pathway by which luteinizing hormone (LH) acts on the somatic cells of vertebrate ovarian follicles to stimulate meiotic resumption in the oocyte requires a decrease in cAMP in the oocyte, but how cAMP is decreased is unknown. Activation of Gi family G proteins can lower cAMP by inhibiting adenylate cyclase or stimulating a cyclic nucleotide phosphodiesterase, but we show here that inhibition of this class of G proteins by injection of pertussis toxin into follicle-enclosed mouse oocytes does not prevent meiotic resumption in response to LH. Likewise, elevation of Ca2+ can lower cAMP through its action on Ca2+-sensitive adenylate cyclases or phosphodiesterases, but inhibition of a Ca2+ rise by injection of EGTA into follicle-enclosed mouse oocytes does not inhibit the LH response. Thus, neither of these well-known mechanisms of cAMP regulation can account for LH signaling to the oocyte in the mouse ovary.


Assuntos
Cálcio/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Hormônio Luteinizante/fisiologia , Meiose/fisiologia , Oócitos/fisiologia , Adenilil Ciclases/metabolismo , Animais , Cálcio/metabolismo , Quelantes/farmacologia , AMP Cíclico/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Técnicas In Vitro , Meiose/efeitos dos fármacos , Camundongos , Oócitos/efeitos dos fármacos , Toxina Pertussis/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa