1
|
Yi ZY, Liang QX, Meng TG, Li J, Dong MZ, Hou Y, Ouyang YC, Zhang CH, Schatten H, Sun QY, Qiao J, Qian WP. PKCβ1 regulates meiotic cell cycle in mouse oocyte. Cell Cycle 2019; 18:395-412. [PMID: 30730241 DOI: 10.1080/15384101.2018.1564492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PKCβI, a member of the classical protein kinase C family, plays key roles in regulating cell cycle transition. Here, we report the expression, localization and functions of PKCβI in mouse oocyte meiotic maturation. PKCβI and p-PKCβI (phosphor-PKCβI) were expressed from germinal vesicle (GV) stage to metaphase II (MII) stage. Confocal microscopy revealed that PKCβI was localized in the GV and evenly distributed in the cytoplasm after GV breakdown (GVBD), and it was concentrated at the midbody at telophase in meiotic oocytes. While, p-PKCβI was concentrated at the spindle poles at the metaphase stages and associated with midbody at telophase. Depletion of PKCβI by specific siRNA injection resulted in defective spindles, accompanied with spindle assembly checkpoint activation, metaphase I arrest and failure of first polar body (PB1) extrusion. Live cell imaging analysis also revealed that knockdown of PKCβI resulted in abnormal spindles, misaligned chromosomes, and meiotic arrest of oocytes arrest at the Pro-MI/MI stage. PKCβI depletion did not affect the G2/M transition, but its overexpression delayed the G2/M transition through regulating Cyclin B1 level and Cdc2 activity. Our findings reveal that PKCβI is a critical regulator of meiotic cell cycle progression in oocytes. Abbreviations: PKC, protein kinase C; COC, cumulus-oocyte complexes; GV, germinal vesicle; GVBD, germinal vesicle breakdown; Pro-MI, first pro-metaphase; MI, first metaphase; Tel I, telophase I; MII, second metaphase; PB1, first polar body; SAC, spindle assembly checkpoint.
Collapse
Affiliation(s)
- Zi-Yun Yi
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Qiu-Xia Liang
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Tie-Gang Meng
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Jian Li
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Ming-Zhe Dong
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Yi Hou
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Ying-Chun Ouyang
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Chun-Hui Zhang
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Heide Schatten
- c Department of Veterinary Pathobiology , University of Missouri-Columbia , Columbia , MO , USA
| | - Qing-Yuan Sun
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Jie Qiao
- d Reproductive Medical Center , Peking University Third Hospital , Beijing , China
| | - Wei-Ping Qian
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| |
Collapse
|
2
|
Herta AC, Lolicato F, Smitz JEJ. In vitro follicle culture in the context of IVF. Reproduction 2018; 156:F59-F73. [PMID: 29980584 DOI: 10.1530/rep-18-0173] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/19/2018] [Indexed: 12/24/2022]
Abstract
The currently available assisted reproduction techniques for fertility preservation (i.e. in vitro maturation (IVM) and in vitro fertilization) are insufficient as stand-alone procedures as only few reproductive cells can be conserved with these techniques. Oocytes in primordial follicles are well suited to survive the cryopreservation procedure and of use as valuable starting material for fertilization, on the condition that these could be grown up to fully matured oocytes. Our understanding of the biological mechanisms directing primordial follicle activation has increased over the last years and this knowledge has paved the way toward clinical applications. New multistep in vitro systems are making use of purified precursor cells and extracellular matrix components and by applying bio-printing technologies, an adequate follicular niche can be built. IVM of human oocytes is clinically applied in patients with polycystic ovary/polycystic ovary syndrome; related knowhow could become useful for fertility preservation and for patients with maturation failure and follicle-stimulating hormone resistance. The expectations from the research on human ovarian tissue and immature oocytes cultures, in combination with the improved vitrification methods, are high as these technologies can offer realistic potential for fertility preservation.
Collapse
Affiliation(s)
- Anamaria C Herta
- Follicle Biology LaboratoryVrije Universiteit Brussel, Brussels, Belgium
| | - Francesca Lolicato
- Follicle Biology LaboratoryVrije Universiteit Brussel, Brussels, Belgium
| | - Johan E J Smitz
- Follicle Biology LaboratoryVrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
3
|
Stricker SA, Cline C, Goodrich D. Oocyte maturation and fertilization in marine nemertean worms: using similar sorts of signaling pathways as in mammals, but often with differing results. THE BIOLOGICAL BULLETIN 2013; 224:137-155. [PMID: 23995739 DOI: 10.1086/bblv224n3p137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
In marine worms belonging to the phylum Nemertea, oocyte maturation and fertilization are regulated by the same general kinds of signals that control such processes in mammals. However, unlike mammalian oocytes that develop within follicles, nemertean oocytes characteristically lack a surrounding sheath of follicle cells and often respond differently to maturation-related cues than do mammalian oocytes. For example, elevators of cyclic adenosine monophosphate (cAMP) or cyclic guanosine monophosphate (cGMP) levels promote the resumption of meiotic maturation (=germinal vesicle breakdown, GVBD) in nemertean oocytes, whereas increasing intraoocytic cAMP and cGMP typically blocks GVBD in mammals. Similarly, AMP-activated kinase (AMPK) signaling keeps nemertean oocytes from maturing, but in mouse oocytes, AMPK activation triggers GVBD. In addition, protein kinase C (PKC) activity is required for seawater-induced GVBD in nemerteans, whereas some PKCs have been shown to inhibit GVBD in mammals. Furthermore, although fertilization causes both types of oocytes to reorganize their endoplasmic reticulum and generate calcium oscillations that can involve soluble sperm factor activity and inositol 1,4,5-trisphosphate signaling, some discrepancies in the spatiotemporal patterns and underlying mechanisms of fertilization are also evident in nemerteans versus mammals. Thus, to characterize differences and similarities in gamete biology more fully, aspects of oocyte maturation and fertilization in marine nemertean worms are reviewed and briefly compared with related findings that have been published for mammalian oocytes. In addition, possible causes of the alternative responses displayed by oocytes in these two animal groups are addressed.
Collapse
Affiliation(s)
- Stephen A Stricker
- Department of Biology, University of New Mexico, Albuquerque, 87131, USA
| | | | | |
Collapse
|
4
|
Lee HS, Kim EY, Lee KA. Changes in gene expression associated with oocyte meiosis after Obox4 RNAi. Clin Exp Reprod Med 2011; 38:68-74. [PMID: 22384421 PMCID: PMC3283059 DOI: 10.5653/cerm.2011.38.2.68] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 05/31/2011] [Accepted: 06/03/2011] [Indexed: 12/03/2022] Open
Abstract
Objective Previously, we found that oocyte specific homeobox (Obox) 4 plays significant role in completion of meiosis specifically at meiosis I-meiosis II (MI-MII) transition. The purpose of this study was to determine the mechanism of action of Obox4 in oocyte maturation by evaluating downstream signal networking. Methods The Obox4 dsRNA was prepared by in vitro transcription and microinjected into the cytoplasm of germinal vesicle oocytes followed by in vitro maturation in the presence or absence of 0.2 mM 3-isobutyl-1-metyl-xanthine. Total RNA was extracted from 200 oocytes of each group using a PicoPure RNA isolation kit then amplified two-rounds. The probe hybridization and data analysis were used by Affymetrix GeneChip® Mouse Genome 430 2.0 array and GenPlex 3.0 (ISTECH, Korea) software, respectively. Results Total 424 genes were up (n=80) and down (n=344) regulated after Obox4 RNA interference (RNAi). Genes mainly related to metabolic pathways and mitogen-activated protein kinase (MAPK) signaling pathway was changed. Among the protein kinase C (PKC) isoforms, PKC-alpha, beta, gamma were down-regulated and especially the MAPK signaling pathway PKC-gamma was dramatically decreased by Obox4 RNAi. In the cell cycle pathway, we evaluated the expression of genes involved in regulation of chromosome separation, and found that these genes were down-regulated. It may cause the aberrant chromosome segregation during MI-MII transition. Conclusion From the results of this study, it is concluded that Obox4 is important upstream regulator of the PKC and anaphase-promoting complex action for maintaining intact germinal vesicle.
Collapse
Affiliation(s)
- Hyun-Seo Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea
| | | | | |
Collapse
|
5
|
Pharmacological analyses of protein kinases regulating egg maturation in marine nemertean worms: a review and comparison with Mammalian eggs. Mar Drugs 2010; 8:2417-34. [PMID: 20948915 PMCID: PMC2953411 DOI: 10.3390/md8082417] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 07/24/2010] [Accepted: 08/20/2010] [Indexed: 01/29/2023] Open
Abstract
For development to proceed normally, animal eggs must undergo a maturation process that ultimately depends on phosphorylations of key regulatory proteins. To analyze the kinases that mediate these phosphorylations, eggs of marine nemertean worms have been treated with pharmacological modulators of intracellular signaling pathways and subsequently probed with immunoblots employing phospho-specific antibodies. This article both reviews such analyses and compares them with those conducted on mammals, while focusing on how egg maturation in nemerteans is affected by signaling pathways involving cAMP, mitogen-activated protein kinases, Src-family kinases, protein kinase C isotypes, AMP-activated kinase, and the Cdc2 kinase of maturation-promoting factor.
Collapse
|
6
|
Stricker SA. Roles of protein kinase C isotypes during seawater-versus cAMP-induced oocyte maturation in a marine worm. Mol Reprod Dev 2009; 76:693-707. [DOI: 10.1002/mrd.20993] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
7
|
Strain difference in the timing of meiosis resumption in mouse oocytes: involvement of a cytoplasmic factor(s) acting presumably upstream of the dephosphorylation of p34cdc2kinase. ZYGOTE 2008. [DOI: 10.1017/s0967199400003774] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryOocytes from eight inbred strains of mice were screened for the timing of germinal vesicle breakdown (GVB)in vitro. This characteristic varied between strains, reaching most extreme values in oocytes from AKR and BALB/c mice (3.1 and 1.6h after release from dibutyryl cAMP block, respectively;p<0.0001). The difference between AKR and BALB/c mice was confirmed in experiments in which GVB was inducedin vivoby stimulation with exogenous gonadotrophins. Analysis of the rate of GVB in hybrids obtained after fusion of nuclear and cytoplamic fragments of oocytes from both strains suggests that the factor responsible for the difference between AKR and BALB/c mice is located in the cytoplasm of the proghase oocytes. Finally, in oocytes from both strains stimulated to resume meiotic maturation with okadaic acid, an inhibitor of protein phosphatases types 1 and 2A the rate of GVB was the same (2.2h and 2.3h for AKR and BALB/c, respectively;p= 0.48). This suggests that the difference between strains is not related to the amount or quality of the pre-MPF (Maturation Promoting Factor) stored in the prophase oocyte, but to the factor(s) acting upstream of the dephosphorylation ofp34cdc2. kinase in the pathway leading to pre-MPF activation.
Collapse
|
8
|
Mondadori RG, Neves JP, Gonçalves PBD. Protein kinase C (PKC) role in bovine oocyte maturation and early embryo development. Anim Reprod Sci 2008; 107:20-9. [PMID: 17646065 DOI: 10.1016/j.anireprosci.2007.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2006] [Accepted: 06/11/2007] [Indexed: 11/29/2022]
Abstract
The aims of the present study were to determine the role of protein kinase C (PKC) on meiotic resumption and its effects on pronuclear formation and cleavage in the bovine. Oocytes were matured in the presence of 0, 1, 10 and 100 nM of phorbol 12-myristate 13-acetate (PMA), to evaluate the percentage of germinal vesicle breakdown. To study pronuclear formation and cleavage, oocytes were randomly distributed in four groups and matured in modified TCM-199 with LH and FSH (negative control); 10% of estrous cow serum (positive control); 100 nM of PMA (treatment); 100 nM of 4alpha-PDD (phorbol ester control). Oocytes were also matured in positive control medium, fertilized and transferred to KSOM with increasing concentrations of a PKC inhibitor. The protein profile and the presence of PKC at the end of maturation period were determined by SDS-PAGE followed by Silver Stain and Western blot, respectively. PMA stimulated meiotic resumption in a concentration-dependent manner. PKC stimulation during oocyte maturation caused an increase in pronuclear formation and did not cause parthenogenetic activation. Inhibitor of PKC (MyrPKC) inhibited cleavage in a dose-dependent and irreversible manner. A protein band around 74 kDa was not detected in PMA-treated oocytes and PKC was not detected by Western blot at the end of the maturation period. In conclusion, meiotic resumption was accelerated and the rate of oocytes with two pronuclei was increased when PKC was activated during oocyte maturation. Moreover, cleavage was inhibited in the presence of PMA.
Collapse
Affiliation(s)
- R G Mondadori
- Animal Reproduction Laboratory, Department of Veterinary Medicine, UPIS-Faculdades Integradas, SEPS 712/912, Brasilia, DF 70390-125, Brazil.
| | | | | |
Collapse
|
9
|
Denys A, Avazeri N, Lefèvre B. The PKC pathway and in particular its β1 isoform is clearly involved in meiotic arrest maintenance but poorly in FSH-induced meiosis resumption of the mouse cumulus cell enclosed oocyte. Mol Reprod Dev 2007; 74:1575-80. [PMID: 17474092 DOI: 10.1002/mrd.20748] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PKC modulators were used to investigate the role of the PKC pathway either on the maintenance of meiotic arrest or on FSH-induced maturation of mouse cumulus cell enclosed oocytes (CEOs). (1) Whereas PKC activation (PMA 8 microM) overcomed clearly the HX-maintained meiotic arrest (83.7 +/- 3.6% vs. 16.1 +/- 10.6% GVBD oocytes), PKC inhibition (Calphostin C 100 nM) did not. On the contrary, it better maintained the meiotic arrest than HX alone. (2) No significant effect of PKC activation or inhibition was observed. (3) HX alone maintained PKCbeta1 in the cytoplasm, whereas FSH and PKC activation induced partly its translocation into the nucleus. The results show that whereas the PKC pathway is clearly involved in maintenance of the meiotic arrest through PKCbeta1, it is not involved in FSH-induced meiosis of CEOs.
Collapse
Affiliation(s)
- Anne Denys
- INSERM Eri-18, Université Paris 13, Bobigny, France
| | | | | |
Collapse
|
10
|
Mishra A, Joy KP. 2-Hydroxyestradiol-17β-induced oocyte maturation in catfish (Heteropneustes fossilis) involves protein kinase C and its interaction with protein phosphatases. Comp Biochem Physiol A Mol Integr Physiol 2006; 144:416-22. [PMID: 16730203 DOI: 10.1016/j.cbpa.2006.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 03/12/2006] [Accepted: 03/23/2006] [Indexed: 11/26/2022]
Abstract
In vitro effects of phorbol 12-myristate 13-acetate (PMA), a protein kinase C (PKC) activator, calphostin C (PKC inhibitor) and okadaic acid [OA, a protein phosphatase (PP; PP1 and PP2A) inhibitor] on 2-hydroxyestradiol-17beta (2-OHE(2))-induced oocyte maturation were investigated in the catfish Heteropneustes fossilis. Incubations of postvitellogenic follicles with PMA or OA alone did not induce oocyte maturation. However, co-incubations with 2-OHE(2) and PMA (0.05, 0.5 and 5 microM) or 2-OHE(2) and OA (0.5, 1.0 or 2.0 microM) increased germinal vesicle breakdown (GVBD) significantly over that of 2-OHE(2). Incubation of follicles with calphostin C elicited varied effects on GVBD, low (0.005 and 0.01 microM) and high (5.0 and 10.0 microM) concentrations did not affect GVBD, but medium concentrations (0.05, 0.1, 0.5, 1.0 and 2.5 microM) stimulated it. The medium concentrations elicited a biphasic stimulatory response with peak GVBD at 0.1 microM (54%). Calphostin C (>or=2.5 microM) inhibited the 2-OHE(2)-induced GVBD in a concentration-dependent manner during the 24 h incubation. Pre- or post-treatment with calphostin C inhibited the steroid-induced GVBD only at 6 h. In co-incubation studies, both PMA and OA reversed the inhibitory effect of calphostin C: the former partially and the latter fully. The results of the present study show that PKC appears to modulate the 2-OHE(2)-induced oocyte maturation. The OA-sensitive PP may be involved in the PKC modulation of steroid-induced oocyte maturation.
Collapse
Affiliation(s)
- Abha Mishra
- Department of Zoology, Banaras Hindu University, Varanasi-221005, India
| | | |
Collapse
|
11
|
Avazeri N, Denys A, Lefèvre B. Lead cations affect the control of both meiosis arrest and meiosis resumption of the mouse oocyte in vitro at least via the PKC pathway. Biochimie 2006; 88:1823-9. [PMID: 16740354 DOI: 10.1016/j.biochi.2006.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 04/20/2006] [Indexed: 10/24/2022]
Abstract
The aim of this study was to determine in vitro whether lead has a direct cytotoxic effect on the female gamete or through its surrounding somatic cells. We had previously demonstrated that it partly accumulates in the mouse ovary and induces follicle and oocyte apoptosis. The data reported here demonstrate for the first time that low levels of Pb(NO3)2 (<or=10 pM) affect oocyte meiosis in vitro. On the one hand, in condition similar to the in vivo one, i.e. in hypoxanthine (HX)-maintained meiotic arrest, Pb(NO3)2 was able to significantly release the oocytes from this arrest. On the other hand, when meiosis occurred spontaneously in vitro, Pb(NO3)2 inhibits meiosis. Whereas PMA, an agonist of PKC was able to prevent the first lead effect, calphostin C, an antagonist, was able to significantly prevent lead inhibition of spontaneous GVBD. And, similarly to Pb2+, the inhibition of PKC by calphostin C prevented HX to maintain the meiotic arrest whereas its activation by PMA inhibited spontaneous GVBD. No significant differences in the effects of Pb(NO3)2 on the oocytes were observed whatever the cumulus cells were present or absent. Moreover, lead did not seem to affect the metaphase plate formation. We concluded that Pb2+ may disturb the control of oocyte meiosis at least in part through its ability to interfere with the PKC pathway in taking place of Ca2+ ions.
Collapse
Affiliation(s)
- N Avazeri
- Institut National de la Santé et de la Recherche Médicale, Unité 566, Commissariat à L'énergie Atomique, Universités de Paris VII et Paris XI, BP 6, 92260 Fontenay-aux-Roses, France
| | | | | |
Collapse
|
12
|
Villaescusa JC, Allard P, Carminati E, Kontogiannea M, Talarico D, Blasi F, Farookhi R, Verrotti AC. Clast4, the murine homologue of human eIF4E-Transporter, is highly expressed in developing oocytes and post-translationally modified at meiotic maturation. Gene 2005; 367:101-9. [PMID: 16343815 DOI: 10.1016/j.gene.2005.09.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 09/23/2005] [Accepted: 09/23/2005] [Indexed: 10/25/2022]
Abstract
In metazoans, translational regulation of a set of maternal mRNAs directs oocyte maturation and early embryogenesis. These transcripts are often kept dormant until their products are spatially and temporally required in development. The interaction between general translation factors (i.e. eIF4E) and their specific interactors influences translation initiation. A search of the protein database for a mouse homologue of the Drosophila Cup protein, a translational repressor during female germ-line development, identified the product of the Clast4 gene. In this report, we show that Clast4 mRNA and protein are highly expressed within the cytoplasm of growing oocytes. The Clast4 protein is stable during this developmental window and post-translationally modified by phosphorylation upon oocyte meiotic maturation. Additionally, we show that Clast4 and eIF4E directly interact by means of a canonical and functional eIF4E-binding motif. Our results suggest that Clast4, similar to Drosophila Cup, may act at the translational level during murine female germ-line development.
Collapse
|
13
|
Michaut MA, Williams CJ, Schultz RM. Phosphorylated MARCKS: A novel centrosome component that also defines a peripheral subdomain of the cortical actin cap in mouse eggs. Dev Biol 2005; 280:26-37. [PMID: 15766745 DOI: 10.1016/j.ydbio.2005.01.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Revised: 12/23/2004] [Accepted: 01/04/2005] [Indexed: 01/13/2023]
Abstract
MARCKS (myristoylated alanine-rich C-kinase substrate) is a major substrate for protein kinase C (PKC), a kinase that has multiple functions during oocyte maturation and egg activation, for example, spindle function and cytoskeleton reorganization. We examined temporal and spatial changes in p-MARCKS localization during maturation of mouse oocytes and found that p-MARCKS is a novel centrosome component based its co-localization with pericentrin and gamma-tubulin within microtubule organizing centers (MTOCs). Like pericentrin, p-MARCKS staining at the MI spindle poles was asymmetric. Based on this asymmetry, we found that one end of the spindle was preferentially extruded with the first polar body. At MII, however, the spindle poles had symmetrical p-MARCKS staining. p-MARCKS also was enriched in the periphery of the actin cap overlying the MI or MII spindle to form a ring-shaped subdomain. Because phosphorylation of MARCKS modulates its actin crosslinking function, this localization suggests p-MARCKS functions as part of the contractile apparatus during polar body emission. Our finding that an activator of conventional and novel PKC isoforms did not increase the amount of p-MARCKS suggested that an atypical isoform was responsible for MARCKS phosphorylation. Consistent with this idea, immunostaining revealed that the staining patterns of p-MARCKS and the active form of the atypical PKC zeta/lambda isoform(s) were very similar. These results show that p-MARCKS is a novel centrosome component and also defines a previously unrecognized subdomain of the actin cap overlying the spindle.
Collapse
Affiliation(s)
- Marcela A Michaut
- Center for Research on Reproduction and Women's Health and Department of Obstetrics and Gynecology, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
14
|
Ma Z, Wang X, Hockman S, Snow EC, Hersh LB. Subcellular localization of nardilysin during mouse oocyte maturation. Arch Biochem Biophys 2005; 434:187-94. [PMID: 15629122 DOI: 10.1016/j.abb.2004.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Revised: 10/18/2004] [Indexed: 11/25/2022]
Abstract
We have previously shown that the peptidase, nardilysin, contains a bipartite nuclear localization signal that permits the enzyme to cycle between the nucleus and cytoplasm. In the present study, we report that nardilysin accumulates in the nucleus of an oocyte as a function of its maturation. Nardilysin is predominantly localized in the cytoplasm of an oocyte when initially placed into culture. The enzyme starts to accumulate in the nucleus within 30 min of in vitro culture. After 3 h, nardilysin is found as a spherical structure surrounded by condensed chromosomal DNA. After 18 h of in vitro culture, it co-localizes with beta-tubulin at the spindle apparatus. Cilostamide, a phosphodiesterase 3A inhibitor that inhibits meiosis, blocks accumulation of nuclear nardilysin. This finding demonstrates that the nuclear entry of nardilysin is tightly controlled in the oocyte. Taken together, these experiments strongly suggest a role for nardilysin in meiosis through its dynamic translocation from cytosol to nucleus, and then to the spindle apparatus.
Collapse
Affiliation(s)
- Zhangliang Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky Medical Center, 800 Rose St., Lexington, KY 40536, USA.
| | | | | | | | | |
Collapse
|
15
|
Zheng ZY, Li QZ, Chen DY, Schatten H, Sun QY. Translocation of phospho-protein kinase Cs implies their roles in meiotic-spindle organization, polar-body emission and nuclear activity in mouse eggs. Reproduction 2005; 129:229-34. [PMID: 15695617 DOI: 10.1530/rep.1.00336] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The protein kinase Cs (PKCs) are a family of Ser/Thr protein kinases categorized into three subfamilies: classical, novel, and atypical. The phosphorylation of PKC in germ cells is not well defined. In this study, we described the subcellular localization of phopho-PKC in the process of mouse oocyte maturation, fertilization, and early embryonic mitosis. Confocal microscopy revealed that phospho-PKC (pan) was distributed abundantly in the nucleus at the germinal vesicle stage. After germinal vesicle breakdown, phospho-PKC was localized in the vicinity of the condensed chromosomes, distributed in the whole meiotic spindle, and concentrated at the spindle poles. After metaphase I, phospho-PKC was translocated gradually to the spindle mid-zone during emission of the first polar body. After sperm penetration and electrical activation, the distribution of phospho-PKC was moved from the spindle poles to the spindle mid-zone. After the extrusion of the second polar body (PB2) phospho-PKC was localized in the area between the oocyte and the PB2. In fertilized eggs, phospho-PKC was concentrated in the pronuclei except for the nucleolus. Phospho-PKC was dispersed after pronuclear envelope breakdown, but distributed on the entire spindle at mitotic metaphase. The results suggest that PKC activation may play important roles in regulating spindle organization and stabilization, polar-body extrusion, and nuclear activity during mouse oocyte meiosis, fertilization, and early embryonic mitosis.
Collapse
Affiliation(s)
- Zhen-Yu Zheng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, People's Republic of China
| | | | | | | | | |
Collapse
|
16
|
Avazeri N, Courtot AM, Lefevre B. Regulation of spontaneous meiosis resumption in mouse oocytes by various conventional PKC isozymes depends on cellular compartmentalization. J Cell Sci 2004; 117:4969-78. [PMID: 15367584 DOI: 10.1242/jcs.01375] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we investigated the spatio-temporal distribution of conventional protein kinases C (cPKC) isoforms PKC-alpha, PKC-betaI, PKC-betaII and PKC-gamma in mouse oocytes. The cPKCs were present in the cytoplasm at the start of the process and migrated to the nucleus (or germinal vesicle) before germinal vesicle breakdown, except for PKC-gamma which remained cytoplasmic. In both compartments, the fully phosphorylated form corresponding to the 'mature' enzyme was revealed for PKC-alpha, PKC-betaI and PKC-betaII. Microinjection of specific antibodies against each isozyme in one or the other cell compartment at different times of the meiotic process, permitted us to observe the following: (1) When located in the cytoplasm at the beginning of the process, PKC-alpha is not implicated in germinal vesicle breakdown, PKC-betaI and PKC-gamma are involved in maintaining the meiotic arrest, and PKC-betaII plays a role in meiosis reinitiation. Furthermore, just before germinal vesicle breakdown, these cytoplasmic cPKCs were no longer implicated. (2) When located in the germinal vesicle, PKC-alpha, PKC-betaI and PKC-betaII are involved in meiosis reinitiation. Our data highlight not only the importance of the nuclear pathways in the cell cycle progression, but also their independence of the cytoplasmic ones. Further investigations are however necessary to discover the molecular targets of these cPKCs to better understand the links with the cell cycle progression.
Collapse
Affiliation(s)
- Nathalie Avazeri
- Institut National de la Santé et de la Recherche Médicale Unité 566 Commissariat à l'Energie Atomique, 92260 Fontenay-aux-Roses CEDEX, France
| | | | | |
Collapse
|
17
|
Bertagnolli A, Gonçalves P, Giometti I, Costa L, Oliveira J, Gonçalves I, Barreto K, Emanuelli I, Borges L. Interação entre células do cumulus e atividade da proteína quinase C em diferentes fases da maturação nuclear de oócitos bovinos. ARQ BRAS MED VET ZOO 2004. [DOI: 10.1590/s0102-09352004000400010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Verificou-se a influência da proteína quinase C (PK-C) no reinício e na progressão da meiose em oócitos bovinos, determinando se as células do cumulus são mediadoras da PK-C na regulação da maturação dos oócitos. Complexos cumulus-oócitos (CCO) e oócitos desnudos (OD), distribuídos aleatoriamente em seis tratamentos (T) com base na presença de um ativador da PK-C (PMA) (T1 e T2), de um forbol éster incapaz de ativar a PK-C (4alfa-PDD-controle) (T3 e T4) ou de apenas o meio básico (TCM-199-controle) (T5 e T6), foram cultivados por 7, 9, 12, 18 e 22 horas. A percentagem de rompimento da vesícula germinativa no grupo cultivado com PMA foi maior do que nos dois grupos controle, com e sem células do cumulus. O cultivo de CCO e OD por 12 e 18 horas demonstrou que a PK-C influencia a progressão para os estádios de metáfase I (MI) e metáfase II (MII) de maneira dependente das células do cumulus. Nos períodos de 9 e 22 horas, não foi possível observar diferença entre os grupos quanto aos diferentes estádios de maturação. A ativação da PK-C acelera o reinício da meiose independentemente das células somáticas e acelera a progressão até os estádios de MI e MII na dependência das células do cumulus.
Collapse
|
18
|
Viveiros MM, O'brien M, Eppig JJ. Protein kinase C activity regulates the onset of anaphase I in mouse oocytes. Biol Reprod 2004; 71:1525-32. [PMID: 15229137 DOI: 10.1095/biolreprod.104.031344] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The metaphase-to-anaphase I transition is a key step in the completion of meiosis I. In mouse oocytes, competence to exit metaphase I (MI) is developmentally regulated and typically not acquired until the preovulatory stage. The possible role of protein kinase C (PKC) in regulating this critical transition was assessed in both normal oocytes isolated from small antral follicles (18-day-old B6SJLF1 mice), which have not yet developed the capacity to progress to metaphase II (MII), and also oocytes defective in their ability to exit MI despite development to the preovulatory stage (24-day-old CX8 recombinant inbred strains). In both systems, transient suppression of endogenous PKC activity by treatment with a PKC-specific inhibitor, bisindolylmaleimide I (BIM), promoted the onset of anaphase I in a dose-dependent manner, while activation of PKC with the phorbol ester TPA blocked progression to MII. Following a 2-h incubation with BIM, the majority of oocytes progressed to, and arrested at, MII. The resulting MII oocytes were fertilizable in vitro, showing similar cleavage and blastocyst development rates between BIM treated and untreated controls. Transferred embryos resulted in the development of pups to term in both groups. These data demonstrate that PKC plays an important role in regulating the onset of anaphase I in mouse oocytes. Moreover, it is concluded that oocytes isolated from small antral follicles become blocked at MI due to a PKC-mediated signal, suggesting that acquisition of competence to complete meiosis I involves, in part, the control of PKC activity. Similarly, failure to regulate PKC activity at the preovulatory stage likely promotes arrest at MI.
Collapse
Affiliation(s)
- Maria M Viveiros
- Center for Animal Transgenesis and Germ Cell Research, Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania 19348, USA.
| | | | | |
Collapse
|
19
|
Viveiros MM, O'Brien M, Wigglesworth K, Eppig JJ. Characterization of protein kinase C-delta in mouse oocytes throughout meiotic maturation and following egg activation. Biol Reprod 2003; 69:1494-9. [PMID: 12826574 DOI: 10.1095/biolreprod.103.019018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Changes in protein kinase C (PKC) activity influence the progression of meiosis; however, the specific function of the various PKC isoforms in female gametes is not known. In the current study, the protein expression and subcellular distribution profile of PKC-delta (PKC-delta), a novel isoform of the PKC family, was determined in mouse oocytes undergoing meiotic maturation and following egg activation. The full-length protein was observed as a doublet (76 and 78 kDa) on Western blot analysis. A smaller (47 kDa) carboxyl-terminal fragment, presumably the truncated catalytic domain of PKC-delta, was also strongly expressed. Both the full-length protein and the catalytic fragment became phosphorylated coincident with the resumption of meiosis and remained phosphorylated throughout metaphase II (MII) arrest. Immunofluorescence staining showed PKC-delta distributed diffusely throughout the cytoplasm of oocytes during maturation and associated with the spindle apparatus during the first meiotic division. Discrete foci of the protein also localized with the chromosomes in some mature eggs. Following the completion of meiosis, PKC-delta became dephosphorylated within 2 h of in vitro fertilization or parthenogenetic activation. The protein also accumulated in the nuclei of early embryos and was phosphorylated during M-phase of the initial mitotic cleavage division. By the two-cell stage, expression of the truncated catalytic fragment was minimal. These data demonstrate that the subcellular distribution and posttranslational modification of PKC-delta is cell cycle dependent, suggesting that its activity and/or function likely vary with the progression of meiosis and egg activation.
Collapse
|
20
|
Affiliation(s)
- Ekaterina Voronina
- Department of Molecular and Cell Biology, Brown University, 69 Brown St, Providence, RI 02912, USA
| | | |
Collapse
|
21
|
Faerge I, Terry B, Kalous J, Wahl P, Lessl M, Ottesen JL, Hyttel P, Grøndahl C. Resumption of meiosis induced by meiosis-activating sterol has a different signal transduction pathway than spontaneous resumption of meiosis in denuded mouse oocytes cultured in vitro. Biol Reprod 2001; 65:1751-8. [PMID: 11717137 DOI: 10.1095/biolreprod65.6.1751] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The sterol 4,4-dimethyl-5-cholesta-8,14,24-trien-3-ol (follicular fluid meiosis-activating sterol [FF-MAS]) isolated from human follicular fluid induces resumption of meiosis in mouse oocytes cultured in vitro. The purpose of this study was to examine the hypothesis that differential signal transduction mechanisms exist for FF-MAS-induced and spontaneous in vitro resumption of meiosis in mouse oocytes. Mouse oocytes were dissected from ovaries originating from mice primed with FSH 48 h before oocyte collection. Mechanically denuded germinal vesicle (GV) oocytes were in vitro matured in medium supplemented with hypoxanthine and FF-MAS or allowed to mature spontaneously; both groups were exposed to individual compounds known to inhibit specific targets in the cell. After 20-22 h of in vitro maturation, resumption of meiosis was assessed as the frequency of oocytes in GV breakdown (GVBD) stage. Pertussis toxin (2.5 microg/ml) did not influence resumption of meiosis in either group. Dibutyryl cyclic GMP (320 microM) inhibited FF-MAS-induced GVBD, but not spontaneous GVBD, whereas the subtype 5 phosphodiesterase-inhibitor zaprinast (50 microM) inhibited GVBD in both groups. Microinjection of the catalytic subunit of cAMP-dependent protein kinase into oocytes inhibited spontaneous GVBD, but not FF-MAS-induced GVBD. An inhibitor of cytoplasmic polyadenylation, cordycepin (80 microM), inhibited or retarded spontaneous GVBD to a further extent than it did FF-MAS-induced GVBD. Spontaneous GVBD was more sensitive to the histone H1 kinase-inhibitor olomoucine (250 microM) than was FF-MAS-induced GVBD. Addition of the mitogen-activated protein kinase (MAPK)-inhibitor PD 98059 (50 microM), phospholipase C-inhibitor U-73122 (10 microM), p21(ras)-inhibitor lovastatine (250 microM), and the src-like kinase inhibitor PP2 (20 microg/ml) inhibited FF-MAS-induced GVBD, but not spontaneous GVBD. Both MAPKs, extracellular regulated kinase (ERK) 1 and ERK2, were phosphorylated under FF-MAS-induced meiotic resumption, in contrast to spontaneous meiotic resumption, in which ERK1 and ERK2 phosphorylation occurred 2 h after GVBD. In the present study, we show that FF-MAS acts through an MAPK-dependent pathway, and we suggest that src-like kinase, p21(ras), and phosphoinositide signaling lie upstream of MAPK in the FF-MAS-activated signaling pathway. Clearly, striking pathway differences are present between spontaneous versus FF-MAS-induced meiotic resumption.
Collapse
Affiliation(s)
- I Faerge
- Fertility Team, SAC 2.02, Novo Nordisk A/S, 2820 Gentofte, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Viveiros MM, Hirao Y, Eppig JJ. Evidence that protein kinase C (PKC) participates in the meiosis I to meiosis II transition in mouse oocytes. Dev Biol 2001; 235:330-42. [PMID: 11437440 DOI: 10.1006/dbio.2001.0311] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oocytes from LTXBO mice exhibit a delayed entry into anaphase I and frequently enter interphase after the first meiotic division. This unique oocyte model was used to test the hypothesis that protein kinase C (PKC) may regulate the meiosis I-to-meiosis II transition. PKC activity was detected in LTXBO oocytes at prophase I and increased with meiotic maturation, with the highest (P < 0.05) activity observed at late metaphase I (MI). Treatment of late MI-stage oocytes with the PKC inhibitor, bisindolylmaleimide I (BIM), transiently reduced (P < 0.05) M-phase-promoting factor (MPF) activity and promoted (P < 0.05) progression to metaphase II (MII), while mitogen-activated protein kinase (MAPK) activity remained elevated during the MI-to-MII transition. Confocal microscopy analysis of LTXBO oocytes during this transition showed PKC-delta associated with the meiotic spindle and then with the chromosomes at MII. Inhibition of PKC activity also prevented untimely entry into interphase, but only when PKC activity was reduced in oocytes before the progression to MII and thus indicates that the transition into interphase is directly associated with the delayed triggering of anaphase I. Moreover, the defect(s) that initiate activation occur upstream of MAPK, as suppression of PKC activity failed to prevent activation by Mos(tm1Ev)/ Mos(tm1Ev) LTXBO oocytes expressing no detectable MAPK activity. In summary, PKC participates in the regulatory mechanisms that delay entry into anaphase I in LTXBO oocytes, and the disruption promotes untimely entry into interphase. Thus, loss of regulatory control over PKC activity during oocyte maturation disrupts the critical MI-to-MII transition, leading to a precocious exit from meiosis.
Collapse
Affiliation(s)
- M M Viveiros
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | |
Collapse
|
23
|
Lu Q, Smith GD, Chen DY, Yang Z, Han ZM, Schatten H, Sun QY. Phosphorylation of mitogen-activated protein kinase is regulated by protein kinase C, cyclic 3',5'-adenosine monophosphate, and protein phosphatase modulators during meiosis resumption in rat oocytes. Biol Reprod 2001; 64:1444-50. [PMID: 11319150 DOI: 10.1095/biolreprod64.5.1444] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Mitogen-activated protein (MAP) kinase, protein kinase C (PKC), cAMP, and okadaic acid (OA)-sensitive protein phosphatases (PPs) have been suggested to be involved in oocyte meiotic resumption. However, whether these protein kinases and phosphatases act by independent pathways or interact with each other in regulating meiosis resumption is unknown. In the present study, we aimed to determine the regulation of meiosis resumption and MAP kinase phosphorylation by PKC, cAMP, and OA-sensitive PPs in rat oocytes using an in vitro oocyte maturation system and Western blot analysis. We found that ERK1 and ERK2 isoforms of MAP kinases existed in a dephosphorylated (inactive) form in germinal vesicle breakdown (GVBD)-incompetent and GVBD-competent germinal vesicle intact (GVI) oocytes as well as GVBD oocytes at equivalent levels. These results indicate that MAP kinases are not responsible for the initiation of normal meiotic resumption in rat oocytes. However, when GVBD-incompetent and GVBD-competent oocytes were incubated in vitro for 5 h, MAP kinases were phosphorylated (activated) in GVBD-competent oocytes, but not in meiotic-incompetent oocytes, suggesting that oocytes acquire the ability to phosphorylate MAP kinase during acquisition of meiotic competence. We also found that both meiosis resumption and MAP kinase phosphorylation were inhibited by PKC activation or cAMP elevation. Moreover, these inhibitory effects were overcome by OA, which inhibited PP1/PP2A activities. These results suggest that both cAMP elevation and PKC activation inhibit meiosis resumption and MAP kinase phosphorylation at a step prior to OA-sensitive protein phosphatases. In addition, inhibitory effects of cAMP elevation on meiotic resumption and MAP kinase phosphorylation were not reversed by calphostin C-induced PKC inactivation, indicating that cAMP inhibits both meiotic resumption and MAP kinase activation in a PKC-independent manner.
Collapse
Affiliation(s)
- Q Lu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, The Chinese Academy of Sciences, Beijing 100080, P.R. China
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
In this study, the possible role of protein kinase C (PKC) in mediating both positive and negative actions on meiotic maturation in isolated mouse oocytes has been examined. When cumulus cell-enclosed oocytes (CEO) were cultured for 17-18 hr in a medium containing 4 mM hypoxanthine (HX) to maintain meiotic arrest, each of the five different activators and five different antagonists of PKC stimulated germinal vesicle breakdown (GVB) in a dose-dependent fashion. One of the activators, phorbol-12-myristate 13-acetate (PMA), also triggered GVB in CEO arrested with isobutylmethylxanthine or guanosine, but not in those arrested with dibutyryl cyclic AMP. When denuded oocytes (DO) were cultured for 3hr in inhibitor-free medium, all PKC activators suppressed maturation (<10% GVB compared to 94% in controls), while the effect of PKC antagonists was negligible. Four of the five antagonists reversed the meiosis-arresting action of HX in DO. PMA transiently arrested the spontaneous maturation of both CEO and DO, with greater potency in DO. The stimulatory action of PMA in HX-arrested oocytes was dependent on cumulus cells, because meiotic induction occurred in CEO but not DO. PKC activators also preferentially stimulated cumulus expansion when compared to antagonists. A cell-cell coupling assay determined that the action of PMA on oocyte maturation was not due to a loss of metabolic coupling between the oocyte and cumulus oophorus. Finally, Western analysis demonstrated the presence of PKCs alpha, beta1, delta, and eta in both cumulus cells and oocytes, but only PKC epsilon was detected in the cumulus cells. It is concluded that direct activation of PKC in the oocyte suppresses maturation, while stimulation within cumulus cells generates a positive trigger that leads to meiotic resumption.
Collapse
Affiliation(s)
- S M Downs
- Biology Department, Marquette University, Milwaukee, WI 53233, USA.
| | | | | |
Collapse
|
25
|
Suprynowicz FA, Groigno L, Whitaker M, Miller FJ, Sluder G, Sturrock J, Whalley T. Activation of protein kinase C alters p34(cdc2) phosphorylation state and kinase activity in early sea urchin embryos by abolishing intracellular Ca2+ transients. Biochem J 2000; 349:489-99. [PMID: 10880348 PMCID: PMC1221172 DOI: 10.1042/0264-6021:3490489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The p34(cdc2) protein kinase, a universal regulator of mitosis, is controlled positively and negatively by phosphorylation, and by association with B-type mitotic cyclins. In addition, activation and inactivation of p34(cdc2) are induced by Ca(2+) and prevented by Ca(2+) chelators in permeabilized cells and cell-free systems. This suggests that intracellular Ca(2+) transients may play an important physiological role in the control of p34(cdc2) kinase activity. We have found that activators of protein kinase C can be used to block cell cycle-related alterations in intracellular Ca(2+) concentration ([Ca(2+)](i)) in early sea urchin embryos without altering the normal resting level of Ca(2+). We have used this finding to investigate whether [Ca(2+)](i) transients control p34(cdc2) kinase activity in living cells via a mechanism that involves cyclin B or the phosphorylation state of p34(cdc2). In the present study we show that the elimination of [Ca(2+)](i) transients during interphase blocks p34(cdc2) activation and entry into mitosis, while the elimination of mitotic [Ca(2+)](i) transients prevents p34(cdc2) inactivation and exit from mitosis. Moreover, we find that [Ca(2+)](i) transients are not required for the synthesis of cyclin B, its binding to p34(cdc2) or its destruction during anaphase. However, in the absence of interphase [Ca(2+)](i) transients p34(cdc2) does not undergo the tyrosine dephosphorylation that is required for activation, and in the absence of mitotic [Ca(2+)](i) transients p34(cdc2) does not undergo threonine dephosphorylation that is normally associated with inactivation. These results provide evidence that intracellular [Ca(2+)](i) transients trigger the dephosphorylation of p34(cdc2) at key regulatory sites, thereby controlling the timing of mitosis entry and exit.
Collapse
Affiliation(s)
- F A Suprynowicz
- Laboratory of Cellular and Molecular Biophysics, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Luria A, Tennenbaum T, Sun QY, Rubinstein S, Breitbart H. Differential localization of conventional protein kinase C isoforms during mouse oocyte development. Biol Reprod 2000; 62:1564-70. [PMID: 10819756 DOI: 10.1095/biolreprod62.6.1564] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Protein kinase C (PKC), the major cell target for tumor-promoting phorbol esters, plays a central role in signal transduction pathways. In many biological systems where Ca(2+) serves as a second messenger, regulatory control is mediated by PKC. The activation of PKC depends on its binding to RACK1 receptor, which is an intracellular protein anchor for activated PKC. We demonstrate that the conventional PKC (cPKC) isoforms, PKC-alpha, PKC-betaI, and PKC-betaII, as well as RACK1, are expressed in mouse oocytes (germinal vesicle [GV]) and mature eggs (metaphase II [MII]). In GV oocytes, PKC-alpha, PKC-betaII, and RACK1 were uniformly distributed in the cytoplasm, while PKC-betaI was localized in the cytoplasm and in the plasma membrane as well. Treatment of GV oocytes with the biologically active phorbol ester, 12-o-tetradecanoyl phorbol-13-acetate (TPA), resulted in a rapid translocation of the cytosolic PKC-alpha, but not PKC-betaI, PKC-betaII, or RACK1, to the plasma membrane. This was associated with inhibition of GV breakdown. In MII eggs (17 h post-hCG), PKC-alpha was uniformly distributed in the cytoplasm while PKC-betaI and -betaII were distributed in the cytoplasm and in the plasma membrane as well. Treatment with TPA resulted in a rapid translocation of PKC-alpha from the cytoplasm to the plasma membrane and a significant decrease of PKC-betaI throughout the cytoplasm, while it also remained in the cell periphery. No change in the distribution of PKC-betaII or RACK1 was observed. TPA also induced pronucleus formation. Physiological activation of MII eggs by sperm induced cortical granule exocytosis associated with significant translocation of PKC-alpha and -betaI, but not -betaII, to the plasma membrane. Overall, these results suggest a possible involvement of cPKC isoforms in the mechanism of mouse oocyte maturation and egg activation.
Collapse
Affiliation(s)
- A Luria
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | | | | | | | | |
Collapse
|
27
|
Su YQ, Xia GL, Byskov AG, Fu GD, Yang CR. Protein kinase C and intracellular calcium are involved in follicle-stimulating hormone-mediated meiotic resumption of cumulus cell-enclosed porcine oocytes in hypoxanthine-supplemented medium. Mol Reprod Dev 1999; 53:51-8. [PMID: 10230816 DOI: 10.1002/(sici)1098-2795(199905)53:1<51::aid-mrd6>3.0.co;2-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The present experiments were conducted to examine the hypothesis that follicle-stimulating hormone (FSH) can stimulate the hydrolysis of phosphoinositide, generating the intracellular second messengers to activate protein kinase C and mobilizing intracellular calcium, thus inducing oocyte meiotic resumption. Pig cumulus cell-enclosed oocytes (CEO) were cultured for 24 hr in 4 mM hypoxanthine (HX)-supplemented medium and treated with different agents in the following designs: (1) CEO were treated with neomycin (an inhibitor of phosphoinositide hydrolysis) in the presence of FSH or only treated with 7,12-dimethylbenzin(a) anthracene (DMBA, a tumor promoter which can cause phosphorylation of phospholipase C (PLC), formation of inositol triphophate, and mobilization of intracellular calcium) to mimic the direct activation of PLC; (2) CEO were challenged by FSH, together with sphingosine or staurosporine (two kinds of PKC inhibitors); or treated with phorbol myristate acetate (PMA, an activator of PKC) separately; (3) CEO were primed with BAPTA/AM (an intracellular calcium chelator) or BAPTA/AM +FSH for 60 min, and then transferred into a new culture medium supplemented with FSH but without BAPTA/AM; total culture time was 24 hr. At the end of the culture, the incidence of germinal vesicle breakdown (GVBD) was calculated. The results showed that: (1) FSH (100 U/liter) could stimulate pig CEO to override the arrest of HX and resume meiosis; DMBA (10(-8)-10(-5) M) itself also had such a kind of effect; whereas neomycin, at the level of 10-20 mM, could dramatically inhibit the stimulatory effect of FSH. (2) Staurosporine (10(-9)-10(-6) M) or sphingosine (10(-8)-10(-5) M) could also inhibit the effect of FSH in a dose-dependent manner on stimulating CEO to resume meiosis. However, PMA (10(-8)-10(-5) M) alone had a dual effect on the meiotic resumption of pig CEO. PMA, at the level of 10(-8)-10(-6) M, could stimulate CEO to resume meiosis, and at high concentration of 10(-5) M , it could even enhance the inhibitory effect of HX. (3) Priming CEO with BAPTA/AM only or BAPTA/AM +FSH for 60 min could significantly inhibit the effect of FSH in a dose-dependent manner. These results indicate that in the process of ligand-mediated meiotic resumption of pig CEO, FSH can stimulate the hydrolysis of phosphoinositide leading to the activation of PKC and mobilization of intracellular calcium; and suggest that multiple signaling pathways and signal interaction are involved in this process.
Collapse
Affiliation(s)
- Y Q Su
- College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
28
|
Sun QY, Rubinstein S, Breitbart H. MAP kinase activity is downregulated by phorbol ester during mouse oocyte maturation and egg activation in vitro. Mol Reprod Dev 1999; 52:310-8. [PMID: 10206663 DOI: 10.1002/(sici)1098-2795(199903)52:3<310::aid-mrd9>3.0.co;2-c] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The effects of protein kinase C (PKC) stimulator, phorbol 12-myriatate 13-acetate (PMA), on meiotic cell cycle regulation and mitogen-activated protein (MAP) kinase changes have been studied in mouse oocytes and eggs. The results showed that MAP kinase activation itself was not necessary for germinal vesicle breakdown (GVBD), but the ability of the ooplasm to phosphorylate MAP kinase was a prerequisite for this event. At concentrations of 1.6 nM, PMA effectively inhibited GVBD and MAP kinase activation, suggesting that PMA inhibits GVBD by inhibiting molecule(s) upstream to MAP kinase. At concentrations of 16.2 nM, PMA induced metaphase-interphase transition more effectively in eggs collected 19 hr after human chorionic gonadotropin (hCG) administration than in those collected 15 hr after hCG administration. The degree of MAP kinase activity decrease was well correlated with the time course and proportion of pronuclear formation. On the other hand, when the effect of PMA on cell cycle progression was abolished by protein phosphatase inhibitor, okadaic acid, MAP kinase was superactivated. The biologically inactive 4 alpha-phorbol 12,13-didecanoate (4 alpha-PDD) had no evident effects on either GVBD and interphase transition or on MAP kinase activity. Furthermore, the effects of PMA on oocyte GVBD, egg activation, and MAP kinase activity could be overcome by the specific PKC inhibitor, calphostin C, suggesting the possible involvement of this enzyme in the regulation of MAP kinase activity. The results suggest that activation of PKC by PMA entrains a cascade of events that ultimately inhibits MAP kinase activation and GVBD in mouse oocytes and induces MAP kinase inactivation and metaphase-interphase transition in mouse eggs.
Collapse
Affiliation(s)
- Q Y Sun
- Department of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | |
Collapse
|
29
|
Coticchio G, Fleming S. Inhibition of phosphoinositide metabolism or chelation of intracellular calcium blocks FSH-induced but not spontaneous meiotic resumption in mouse oocytes. Dev Biol 1998; 203:201-9. [PMID: 9806784 DOI: 10.1006/dbio.1998.9021] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mammalian oocytes are arrested at the diplotene phase of the first meiotic division until ovulation. In the mouse, germinal vesicle breakdown (GVBD) and progression to metaphase II is thought to be triggered by a positive signal originating in the follicular cells following stimulation by the luteinizing hormone (LH) surge. Isolated, fully grown oocytes can also undergo spontaneous reinitiation of meiosis in vitro in the absence of gonadotrophin stimulation. To investigate the mechanism of meiotic resumption, inhibitors of phosphoinositide metabolism and an intracellular calcium chelator were used during maturation in vitro under different conditions. In a series of experiments, isolated cumulus cell-oocyte complexes (COCs) maintained in meiotic arrest by hypoxanthine were induced to resume meiosis by treatment with follicle-stimulating hormone (FSH). Under these conditions, both LiCl and neomycin, which inhibit phosphoinositide hydrolysis, produced a dose-dependent inhibitory effect on meiotic resumption. Similar results were obtained when FSH-induced meiotic resumption was observed in the presence of the acetoxymethyl ester form of 1, 2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA/AM), an intracellular calcium chelator. In hypoxanthine-arrested oocytes, GVBD induced by epidermal growth factor (EGF), which mimics FSH action in in vitro maturation, was also repressed by LiCl and neomycin. Conversely, meiotic resumption triggered by a pulse of 8-bromo-cyclic adenosine monophosphate (8-Br cAMP) was not affected by these two inhibitors. In experiments in which oocytes were cultured under conditions which permit spontaneous meiotic maturation, resumption of meiosis was not affected by either inhibition of phosphoinositide hydrolysis or chelation of intracellular calcium. Therefore, it appears that meiotic resumption induced by hormone stimulation requires activation of the phosphoinositide pathway and mobilization of intracellular calcium. In contrast, spontaneous maturation probably occurs through a different mechanism because it is not affected by inhibition of this signaling pathway.
Collapse
Affiliation(s)
- G Coticchio
- Academic Division of Obstetrics and Gynaecology, School of Human Development, University of Nottingham, Nottingham, United Kingdom.
| | | |
Collapse
|
30
|
Wassarman PM, Florman HM. Cellular Mechanisms During Mammalian Fertilization. Compr Physiol 1997. [DOI: 10.1002/cphy.cp140124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Abstract
To test the hypothesis that culture conditions influence meiotic regulation in mouse oocytes, we have examined the effects of six culture media, four organic buffers, and pH on spontaneous maturation, the maintenance of meiotic arrest and ligand-induced maturation in cumulus cell-enclosed oocytes from hormonally primed immature mice. The media tested were Eagle's minimum essential medium (MEM), Ham's F-10 (F-10), M199, M16, Waymouth's MB 752/1 (MB 752/1), and Leibovitz's L-15 (L-15). All six media supported > or = 94% spontaneous germinal vesicle breakdown (GVB) during a 17-18 hr incubation period, but polar body formation was lower in M199 and MB 752/1 than in the other media. The incidence of polar bodies could be increased in these two media by the addition of pyruvate. With the exception of M16 and MB 752/1, 4 mM hypoxanthine maintained a significant number of cumulus cell-enclosed oocytes in meiotic arrest. Inhibition could be restored by the addition of glutamine to M16 and pyruvate to MB 752/1. Follicle-stimulating hormone (FSH) and epidermal growth factor (EGF) stimulated GVB in those media in which hypoxanthine was inhibitory. dbcAMP was able to maintain meiotic arrest in all of the media, but was least effective in M16. FSH stimulated GVB in all dbcAMP-arrested groups except L-15, and FSH became stimulatory in L-15 when the pyruvate level was reduced to 0.23 mM and galactose was replaced with 5.5 mM glucose. When MEM was buffered principally with the organic buffers MOPS, HEPES, DIPSO, or PIPES (at 20 mM), high frequencies of GVB and polar body formation were observed in inhibitor-free medium. dbcAMP suppressed GVB in all groups; hypoxanthine also maintained meiotic arrest in all buffering conditions, although this effect was nominal in PIPES-buffered medium. FSH and EGF stimulated GVB in all dbcAMP- and hypoxanthine-treated groups. When the concentration of HEPES was increased from 20 mM to 25 mM, a more pronounced suppressive effect on maturation in both dbcAMP- and hypoxanthine-supplemented groups was observed in the absence of FSH. But whereas HEPES reduced the induction of maturation by FSH in dbcAMP-arrested oocytes, this buffer had no effect on FSH action in hypoxanthine-treated oocytes. When MEM was buffered with HEPES and the pH was adjusted to 6.8, 7.0, 7.2, or 7.4, a dramatic effect of pH on meiotic maturation was observed. pH had no significant effect on hypoxanthine salvage by oocyte-cumulus cell complexes, but FSH-induced de novo purine synthesis was significantly augmented by increased pH, in parallel with increased induction of GVB. The results of this study demonstrate that the use of different culture media, or minor changes in culture conditions, can lead to significant variation in (1) the spontaneous maturation of oocytes, (2) the ability of meiotic inhibitors to suppress GVB, or (3) the efficacy of meiosis-inducing ligands. Furthermore, such observations provide a unique opportunity to examine specific molecules and metabolic pathways that can account for this variation and thereby gain valuable insights into the mechanisms involved in meiotic regulation.
Collapse
Affiliation(s)
- S M Downs
- Biology Department, Marquette University, Milwaukee, WI 53233, USA
| | | |
Collapse
|
32
|
Rose-Hellekant TA, Bavister BD. Precocious oocyte maturation is induced by an inhibitor of cAMP-dependent protein kinase in the intact golden hamster. Mol Reprod Dev 1996; 44:250-5. [PMID: 9115724 DOI: 10.1002/(sici)1098-2795(199606)44:2<250::aid-mrd15>3.0.co;2-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The purpose of this investigation was to determine if precocious oocyte maturation could be induced by modulating ovarian cAMP-dependent protein kinase (PKA) or protein kinase C (PKC) signal transduction pathways in the intact hamster. The following inhibitors and stimulators were injected into the ovarian bursal cavity of the anesthetized hamster: N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (H-89), a relatively selective inhibitor of PKA phosphorylations; a structurally related compound, H-7, a less potent and selective inhibitor used to alter PKA and PKC pathways; phorbol 12, 13-didecanoate (PDD beta), an active stimulator of PKC and the inactive analog, 4 alpha-phorbol 12, 13-didecanoate (PDD alpha); and GF109203x, a potent and selective inhibitor of PKC phosphorylations. The experimental design was to inject the modulator into the bursal cavity of one ovary and control solution of diluent or inactive compound into the contralateral bursal cavity. After 1 hr oocytes were collected and evaluated microscopically for the presence or absence of a germinal vesicle. Only oocytes recovered from H-89 treated ovaries (> 50 microM) showed significantly greater frequency of meiotic resumption. Exposure of ovaries to H-7 (< or = 150 microM), PDD beta (< or = 100 microM), or GF109203x (< or = 100 microM) did not significantly affect oocyte maturation state. These results suggest that ovarian protein phosphorylations carried out by PKA are necessary for the maintenance of oocyte meiotic arrest in situ.
Collapse
Affiliation(s)
- T A Rose-Hellekant
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison 53706, USA
| | | |
Collapse
|
33
|
Rose-Hellekant TA, Bavister BD. Roles of protein kinase A and C in spontaneous maturation and in forskolin or 3-isobutyl-1-methylxanthine maintained meiotic arrest of bovine oocytes. Mol Reprod Dev 1996; 44:241-9. [PMID: 9115723 DOI: 10.1002/(sici)1098-2795(199606)44:2<241::aid-mrd14>3.0.co;2-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Four hypotheses were tested using isolated bovine oocytes. (1) Cumulus oocyte complexes (COCs) or denuded oocytes (DOs) were cultured with the protein kinase A (PKA) inhibitor, H-89, to test if meiotic arrest induced by forskolin or IBMX was due to cAMP-stimulated PKA activity or nonspecific effects of these cAMP elevators. (2) COCs were cultured with a protein kinase C (PKC) stimulator (PDD beta) or inhibitor (GF109203x) to test if PKC modulation altered oocyte maturation. (3) COCs were prestimulated for 15 min with (a) PDD beta followed by cotreatment with forskolin, or (b) with H-89 or H-7 followed by cotreatment with GF109203x, to test for interaction between the PKA and PKC signal transduction pathways. (4) H-89 was added to spontaneously maturing COCs at intervals of 0-18 hr to test if H-89 interfered with the transition between meiosis I and II. The results were as follows: H-89 interfered with forskolin or IBMX arrested oocytes in dose-response manner (IBMX ED50 = 41 microM for COCs; forskolin ED50 = 9 microM for denuded oocytes). Prestimulation with PKC induced meiotic resumption in COCs in spite of the presence of forskolin [PDD beta followed by PDD beta + forskolin: 41-47% germinal vesicle (GV) oocytes; forskolin alone: 90-95% GV], while PKC inhibition induced meiotic arrest to a similar extent as forskolin (GF109203x, 85% GV; forskolin, 67-80% GV). Additionally, pretreatment of COCs with H-89 interfered with GF109203x induced arrest (41% vs. 90% GV, respectively). Finally, H-89 interfered with the timely progression of COCs from meiosis I and II. These results indicate that the PKA and PKC pathways can modulate the maturation of bovine oocytes in vitro.
Collapse
Affiliation(s)
- T A Rose-Hellekant
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison 53706, USA
| | | |
Collapse
|
34
|
Abstract
The role of calcium in the regulation of both the meiotic and mitotic cell cycles has been the subject of considerable investigation in the nonmammalian field. In contrast, the mechanisms for signalling meiotic maturation in the mammalian oocyte are not as well documented nor as clearly defined. In the mammalian oocyte, calcium is associated with both spontaneous and hormone-induced meiotic maturation. A transient release of endogenously stored calcium precedes germinal vesicle breakdown and can override cyclic AMP maintained meiotic arrest; it thus may signal the resumption of meiosis. Additionally, extracellular calcium is apparently required for meiotic progression past metaphase I. The time sequence for meiotic resumption and progression is very varied between species. The timing of cell cycle protein synthesis during meiosis suggests that cyclins may be expressed in oocytes of some species much earlier in their development than in others. A generic model is proposed for the mechanism for triggering meiotic resumption in the mammalian oocyte. In this model, the critical components of meiotic resumption involve the temporal relationship of cyclin synthesis and the subsequent activation of the MPF complex by the calcium signal generated, which accounts for differences among species.
Collapse
Affiliation(s)
- S T Homa
- Department of Obstetrics and Gynaecology, St. Bartholomew's Hospital, West Smithfield, London, England
| |
Collapse
|
35
|
Pesty A, Lefèvre B, Kubiak J, Géraud G, Tesarik J, Maro B. Mouse oocyte maturation is affected by lithium via the polyphosphoinositide metabolism and the microtubule network. Mol Reprod Dev 1994; 38:187-99. [PMID: 8080648 DOI: 10.1002/mrd.1080380210] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The incubation of mechanically denuded mouse oocytes in medium containing LiCl delayed both germinal vesicle breakdown (GVBD) and polar body extrusion in a dose-dependent and reversible manner. When myo-inositol alone was added to the culture medium, we observed that it accelerated GVBD and increased the rate of polar body extrusion, whereas, when combined with LiCl, the normal timing of GVBD was recovered. In the same way, when inositol trisphosphate (InsP3) was microinjected into the ooplasma, we observed an important improvement of the rate of GVBD, as compared to control oocytes, and prevention of lithium inhibition. However, neither myo-inositol nor InsP3 were able to rescue totally the oocytes from the negative effect of lithium on polar body extrusion. Moreover, lithium induced some important changes in microtubule and chromosome organizations. Before extrusion of the first polar body, the reduction of the spindle size or the appearance of short individualized chromosomes dispersed around a large aster of microtubules were often observed, whereas, after polar body extrusion, the spindle appeared smaller and chromosomes were often trapped in the midbody. Thus lithium affects mouse oocyte maturation at two different levels: GVBD and polar body extrusion. Whereas the former seems to be affected via polyphosphoinositide turnover, the latter is InsP3-independent and seems to be influenced negatively via underdevelopment of microtubular structures.
Collapse
Affiliation(s)
- A Pesty
- I.N.S.E.R.M., Clamart, France
| | | | | | | | | | | |
Collapse
|
36
|
Dyban AP, De Sutter P, Verlinsky Y. Okadaic acid induces premature chromosome condensation reflecting the cell cycle progression in one-cell stage mouse embryos. Mol Reprod Dev 1993; 34:402-15. [PMID: 8385966 DOI: 10.1002/mrd.1080340409] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Haploid parthenogenetic embryos as well as fertilized mouse eggs were treated in vitro with 1-10 microM okadaic acid (OA) at the one-cell stage. Cytogenetic analysis detected that OA induces nuclear envelope breakdown (NEBD) and premature condensation of interphase chromosomes in pronuclei as well as in 2nd polar body (PB) nuclei. G1-, S-, and G2-type prematurely condensed chromosomes (PCC) were found in pronuclei of embryos of different age, which reflects their progression through the first cell cycle. In nuclei from 2nd PBs only G1- and S-type PCC were observed. Using the types of PCC as a criterion of different phases of the cell cycle, it was possible to estimate that in haploid parthenogenetic embryos G1-phase lasts until 5.5 hr post activation (hpa), S-phase takes from 4.5 to 9.5 hpa, and from 8.5 hpa G2-phase had started. Second PBs were found to be in G1-phase until 6.5 hpa and S-phase started in some as early as 5.5 hpa, but in most not before 7.5 hpa. Treatment with OA visualizes G1-chromosomes in pronuclei as well as in 2nd PBs, and it is easy to count the number of these chromosomes and recognize a T6 marker chromosome. The possibility to apply cytogenetic analysis of G1-chromosomes from 2nd PBs for a more accurate detection of maternal meiotic nondisjunction is discussed.
Collapse
Affiliation(s)
- A P Dyban
- Reproductive Genetics Institute, Illinois Masonic Medical Center, Chicago 60657
| | | | | |
Collapse
|
37
|
Murnane JM, DeFelice LJ. Electrical maturation of the murine oocyte: an increase in calcium current coincides with acquisition of meiotic competence. ZYGOTE 1993; 1:49-60. [PMID: 8081801 DOI: 10.1017/s0967199400001295] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have used the whole-cell recording technique to compare three stages of primary and secondary oocytes from F1 hybrid mice (C57BL/6J x SJL/J): neonatal germinal vesicle (NGV) stage primary oocytes from 10- to 20-day-old, prepubescent mice; mature germinal vesicle (MGV) stage primary oocytes from 12-week-old, post-pubescent, superovulated mice; first polar body (FPB) stage secondary oocytes from 12-week-old, post-pubescent mice during the normal oestrus cycle or following superovulation. NGV, MGV and FPB oocytes all exhibit two voltage-dependent currents: an inward, rapidly activating/inactivating current, and an outward, slowly activating/non-inactivating current. In 1.5 mmol/l external Ca the average peak inward current is -2.9, -12.4 and -13.8 microA/cm2 in NGV, MGV and FPB oocytes, respectively. In 20 mmol/l Ca these currents increase and the reversal potential shifts to the right. The outward current decreases slightly with growth and development: at 40 mV test potentials, NGV oocytes have average outward currents of 8.9 microA/cm2, and MGV and FPB oocytes have currents of 5.0 and 5.5 microA/cm2, respectively. Thus, MGV oocytes express FPB current patterns. The reversal potentials, kinetics and pharmacology of the currents indicate that Ca channels carry the inward current and K channels carry the outward current. During growth in vivo a gradual depolarisation accompanies maturation. Resting potentials ranged from -45 to -30 mV in NGV oocytes to -35 to -17 mV in MGV oocytes to -20 mV to -3 mV in FPB oocytes. These data suggest that a selective increase occurs in the number of Ca channels during oocyte growth. This increase precedes nuclear maturation and coincides with the acquisition of meiotic competence.
Collapse
Affiliation(s)
- J M Murnane
- Emory University School of Medicine, Atlanta, Georgia 30322
| | | |
Collapse
|
38
|
Lefèvre B, Pesty A, Koziak K, Testart J. Protein kinase C modulators influence meiosis kinetics but not fertilizability of mouse oocytes. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1992; 264:206-13. [PMID: 1431782 DOI: 10.1002/jez.1402640213] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The role of protein kinase C (PKC) in the successive steps of mouse oocyte meiotic process was investigated. We have used either OAG, an analog of diacylglycerol, or mezerein, a nonphorbol ester diterpene, less tumor promoting than phorbol esters, as PKC activators, and staurosporine as PKC inhibitor. Cumulus-free oocytes were cultured in minimum essential medium with each of these PKC modulators and maturation stages were screened every two hours until the end of the process. Both PKC activators prevented GVBD at each tested dose for 4 hr (OAG) and 8 hr (mezerein), and decreased the frequencies of PB oocytes. The inhibitory effects of both activators were dose dependent and reversible. The addition of OAG to the culture medium after GVBD occurrence (i.e., after 4 hrs) did not affect PB extrusion whereas similar addition of mezerein significantly decreased the frequency of PB oocytes. Inhibition of PKC by staurosporine accelerated GVBD and increased the frequency of PB extrusion. When staurosporine was added after GVBD, PB extrusion occurred earlier but PB oocyte frequency was not increased. Fertilizability was not affected when oocyte maturation occurred in the presence of any of these substances despite the delay in maturation process. These results clearly indicate that the PKC pathway is involved in mouse oocyte meiotic process: activation of the enzyme would arrest meiotic process whereas its inhibition would participate in meiosis induction.
Collapse
|
39
|
Schwartz DA, Schultz RM. Zygotic gene activation in the mouse embryo: involvement of cyclic adenosine monophosphate-dependent protein kinase and appearance of an AP-1-like activity. Mol Reprod Dev 1992; 32:209-16. [PMID: 1323305 DOI: 10.1002/mrd.1080320305] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protein phosphorylation catalyzed by the cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA) is implicated in regulating zygotic gene activation in the two-cell mouse embryo (Poueymirou and Schultz; Dev Biol 133:588-599, 1989). We now provide evidence that H8, which is a PKA inhibitor, inhibits expression of an hsp70-driven beta-galactosidase reporter gene and that the concentration-dependence of this inhibition is similar to that for inhibiting expression of a stage-specific gene(s) that is a product of zygotic gene activation. We also demonstrate that neither cAMP nor serum can stimulate the expression, as detected by a histochemical assay, of a cAMP response element (CRE)- or serum response element (SRE)-driven beta-galactosidase reporter gene, respectively, in either germinal vesicle-intact oocytes or aphidicolin-arrested one-cell embryos that are chronologically at the tw-cell stage. In contrast, although 12-O-tetradecanoyl phorbol-13-acetate (TPA) does not stimulate expression of a TPA response element (TRE)-driven beta-galactosidase reporter gene in germinal vesicle-intact oocytes, it stimulates such expression in aphidicolin-arrested one-cell embryos. Moreover, TPA can stimulate the expression of either a CRE- or an SRE-driven beta-galactosidase reporter gene in such embryos. Results of these studies further implicate protein phosphorylation in regulating zygotic gene activation, along with its role in modulating enhancer function in the early mouse embryo.
Collapse
Affiliation(s)
- D A Schwartz
- Department of Biology, University of Pennsylvania, Philadelphia 19104-6018
| | | |
Collapse
|
40
|
Stith BJ, Goalstone ML, Kirkwood AJ. Protein kinase C initially inhibits the induction of meiotic cell division in Xenopus oocytes. Cell Signal 1992; 4:393-403. [PMID: 1419482 DOI: 10.1016/0898-6568(92)90034-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have used one activator and two inhibitors of protein kinase C (PKC) to examine the role of this enzyme in the induction of meiotic cell division. At 1 U/ml, phosphatidylcholine-specific phospholipase C increases DAG, alters intracellular pH and inhibits the induction of meiosis by insulin or progesterone. However, when added about 1.6 h after progesterone, the enzyme speeds the induction of cell division. Microinjection of inhibitor peptide (19-36) of PKC has little effect on progesterone action but stimulates the induction of meiosis by insulin. When the inhibitor peptide is injected about 2h after insulin addition, the peptide inhibits. A second PKC inhibitor, staurosporine, decreases PKC-dependent intracellular pH and in vitro oocyte PKC activity. At similar concentrations, staurosporine stimulates insulin or progesterone action, but, when added after about 2 h, the drug inhibits induction by insulin. We conclude that PKC is initially inhibitory to the induction of meiotic cell division but then may become synergistic.
Collapse
Affiliation(s)
- B J Stith
- Department of Biology, University of Colorado, Denver 80217-3364
| | | | | |
Collapse
|
41
|
Wickramasinghe D, Albertini DF. Centrosome phosphorylation and the developmental expression of meiotic competence in mouse oocytes. Dev Biol 1992; 152:62-74. [PMID: 1378414 DOI: 10.1016/0012-1606(92)90156-b] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Previous studies suggested that the transition from an incompetent to a competent meiotic state during the course of oogenesis in the mouse involved a G2/M-like cell cycle transition (Wickramasinghe et al, 1991. Dev. Biol. 143, 162). The present studies tested the hypothesis that centrosome phosphorylation, an event normally induced by MPF, is required for this developmental transition and the expression of meiotic competence in cultured growing mouse oocytes. Multiple fluorescence labeling techniques were used to evaluate centrosome number, phosphorylation status, and microtubule nucleating capacity in competent and incompetent oocytes. Experimental conditions were established for reversibly altering the phosphorylation status of the centrosomes and the effects of these treatments on meiotic resumption were examined. Phosphorylated centrosomes nucleating short microtubules were observed in competent oocytes, whereas nonphosphorylated centrosomes and interphase microtubule arrays were found in incompetent oocytes. Upon recovery from nocodazole-induced microtubule depolymerization, short microtubules formed from centrosomes in competent oocytes, whereas long microtubules reappear in the cytoplasm of incompetent oocytes. Perturbation of the phosphorylation state of oocytes with activators of protein kinase A or protein kinase C resulted in the formation of long interphase microtubules in competent oocytes while centrosome phosphorylation was maintained. Treatment of competent oocytes with the phosphorylation inhibitor 6-dimethylaminopurine also led to formation of long microtubules, although under these conditions centrosomes were dephosphorylated. When competent oocytes were treated simultaneously with puromycin and the phosphodiesterase inhibitor isobutyl methylxanthine (IBMX) for 6 hr, centrosomes became dephosphorylated; centrosomes were rephosphorylated when competent oocytes were further cultured in IBMX without puromycin. Conditions that induced centrosome dephosphorylation in competent oocytes resulted in the loss of the ability to express meiotic competence in culture, whereas maintenance of centrosome phosphorylation in these oocytes was correlated with the ability to resume meiosis. These results suggest that the G2/M transition that occurs when mouse oocytes progress from an incompetent to a competent state in vivo involves the phosphorylation of centrosomes and that the maintenance of centrosome phosphorylation is required for the in vitro expression of meiotic competence.
Collapse
Affiliation(s)
- D Wickramasinghe
- Department of Anatomy and Cell Biology, Tufts University Health Science School, Boston, Massachusetts 02111
| | | |
Collapse
|
42
|
Downs SM, Buccione R, Eppig JJ. Modulation of meiotic arrest in mouse oocytes by guanyl nucleotides and modifiers of G-proteins. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1992; 262:391-404. [PMID: 1320658 DOI: 10.1002/jez.1402620405] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Guanyl nucleotide binding-proteins, or G-proteins, are ubiquitous molecules that are involved in cellular signal transduction mechanisms. Because a role has been established for cAMP in meiosis and G-proteins participate in cAMP-generating systems by stimulating or inhibiting adenylate cyclase, the present study was conducted to examine the possible involvement of G-proteins in the resumption of meiotic maturation. Cumulus cell-free mouse oocytes (denuded oocytes) were maintained in meiotic arrest in a transient and dose-dependent manner when microinjected with the nonhydrolyzable GTP analog, GTP gamma S. This effect was specific for GTP gamma S, because GppNHp, GTP, and ATP gamma S were without effect. Three compounds, known to interact with G-proteins, were tested for their ability to modulate meiotic maturation: pertussis toxin, cholera toxin, and aluminum fluoride (AlF4-). Pertussis toxin had little effect on maturation in either cumulus cell-enclosed oocytes or denuded oocytes when meiotic arrest was maintained with dibutyryl cAMP (dbcAMP) or hypoxanthine. Cholera toxin stimulated germinal vesicle breakdown (GVB) in cumulus cell-enclosed oocytes during long-term culture, but its action was inhibitory in denuded oocytes. AlF4- stimulated GVB in both cumulus cell-enclosed oocytes and denuded oocytes when meiotic arrest was maintained with hypoxanthine but was much less effective in dbcAMP-arrested oocytes. In addition, AlF4- abrogated the inhibitory action of cholera toxin in denuded oocytes and also that of follicle-stimulating hormone (FSH) in cumulus cell-enclosed oocytes. Cholera toxin or FSH alone each stimulated the synthesis of cAMP in oocyte-cumulus cell complexes, whereas pertussis toxin or AlF4- alone were without effect. Both cholera toxin and AlF4- augmented the stimulatory action of FSH on cAMP. These data suggest the involvement of guanyl nucleotides and G-proteins in the regulation of GVB, although different G-proteins and mediators may be involved at the oocyte and cumulus cell levels. Cholera toxin most likely acts by ADP ribosylation of the alpha subunit of Gs and increased generation of cAMP, whereas AlF4- appears to act by antagonizing a cAMP-dependent step.
Collapse
Affiliation(s)
- S M Downs
- Biology Department, Marquette University, Milwaukee, Wisconsin 53233
| | | | | |
Collapse
|
43
|
Carroll J, Swann K. Spontaneous cytosolic calcium oscillations driven by inositol trisphosphate occur during in vitro maturation of mouse oocytes. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(19)49895-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
44
|
Tombes RM, Simerly C, Borisy GG, Schatten G. Meiosis, egg activation, and nuclear envelope breakdown are differentially reliant on Ca2+, whereas germinal vesicle breakdown is Ca2+ independent in the mouse oocyte. J Cell Biol 1992; 117:799-811. [PMID: 1577859 PMCID: PMC2289470 DOI: 10.1083/jcb.117.4.799] [Citation(s) in RCA: 196] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During early development, intracellular Ca2+ mobilization is not only essential for fertilization, but has also been implicated during other meiotic and mitotic events, such as germinal vesicle breakdown (GVBD) and nuclear envelope breakdown (NEBD). In this study, the roles of intracellular and extracellular Ca2+ were examined during meiotic maturation and reinitiation at parthenogenetic activation and during first mitosis in a single species using the same methodologies. Cumulus-free metaphase II mouse oocytes immediately resumed anaphase upon the induction of a large, transient Ca2+ elevation. This resumption of meiosis and associated events, such as cortical granule discharge, were not sensitive to extracellular Ca2+ removal, but were blocked by intracellular Ca2+ chelators. In contrast, meiosis I was dependent on external Ca2+; in its absence, the formation and function of the first meiotic spindle was delayed, the first polar body did not form and an interphase-like state was induced. GVBD was not dependent on external Ca2+ and showed no associated Ca2+ changes. NEBD at first mitosis in fertilized eggs, on the other hand, was frequently, but not always associated with a brief Ca2+ transient and was dependent on Ca2+ mobilization. We conclude that GVBD is Ca2+ independent, but that the dependence of NEBD on Ca2+ suggests regulation by more than one pathway. As cells develop from Ca(2+)-independent germinal vesicle oocytes to internal Ca(2+)-dependent pronuclear eggs, internal Ca2+ pools increase by approximately fourfold.
Collapse
Affiliation(s)
- R M Tombes
- Laboratory of Molecular Biology, University of Wisconsin, Madison 53706
| | | | | | | |
Collapse
|
45
|
Tesarik J, Lefèvre B, Testart J. Progression of oocyte maturation from metaphase I to metaphase II is disturbed by previous immunological interference with cumulus cell function. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1991; 260:116-24. [PMID: 1791418 DOI: 10.1002/jez.1402600115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The effects of an antibody preparation reacting with preovulatory mouse cumuli oophori (anticumulus Ig) on oocyte maturation in vivo and in vitro were studied. Continuous presence of anticumulus Ig in culture medium did not impair oocyte maturation in vitro. Similarly, no effect on oocyte maturation in vivo was observed when anticumulus Ig was given to females superovulated with pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) at the time of hCG treatment. However, when administered earlier, anticumulus Ig brought about serious disturbances of oocyte meiotic competence, since only immature oocytes were ovulated after anticumulus Ig injection at the time of PMSG treatment and as much as 70% of the ovulated oocytes were immature when the antibody was applied 24 hr later. Previous absorption of anticumulus Ig with isolated cumulus cells removed the inhibitory effect of this preparation on oocyte meiotic competence to the same extent as absorption with whole cumuli oophori, despite the persistence of a strong reactivity of the cumulus cell-absorbed antibody preparations with the cumulus intercellular matrix. The ability of oocytes obtained from antibody-injected animals to mature in vitro was also considerably impaired when the injection was made at the time of PMSG treatment. In all cases the maturation defect concerned the progression of meiosis from metaphase I to metaphase II, while the ability of oocytes to undergo germinal vesicle breakdown (GVBD) was unchanged.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- J Tesarik
- Institut National de la Santé, Unité 187, Clamart, France
| | | | | |
Collapse
|
46
|
Kulyk WM. Promotion of embryonic limb cartilage differentiation in vitro by staurosporine, a protein kinase C inhibitor. Dev Biol 1991; 146:38-48. [PMID: 2060709 DOI: 10.1016/0012-1606(91)90444-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Phorbol 12-myristate 13-acetate (PMA), a protein kinase C-activating phorbol ester, is known to inhibit chondrogenic differentiation by embryonic limb mesenchyme cells in vitro. The present study demonstrates that staurosporine, a potent inhibitor of protein kinase C, conversely stimulates cartilage differentiation in cultures of limb mesenchyme cells isolated from whole wing buds of stage 23/24 chick embryos or from the distal subridge region of stage 25 wing buds. In high density micromass cultures, in which limb mesenchyme cells undergo extensive spontaneous cartilage differentiation, exposure to 5-20 nM staurosporine promotes an accelerated accumulation of type II collagen and cartilage proteoglycan mRNA transcripts and a 2- to 3-fold increase in matrix glycosaminoglycan deposition. Even in low density, monolayer cultures in which the mesenchymal cells do not normally form cartilage, treatment with 5 nM staurosporine induces extensive Alcian blue-positive matrix production, a striking 4- to 18-fold rise in sulfated glycosaminoglycan accumulation, and a dramatic elevation of cartilage-characteristic gene transcript expression. Moreover, concurrent treatment with staurosporine overcomes the inhibitory effects of PMA on in vitro limb cartilage differentiation. The results suggest the hypothesis that protein kinase C might function as a negative modulator of chondrogenic differentiation during embryonic limb development.
Collapse
Affiliation(s)
- W M Kulyk
- Department of Anatomy, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
47
|
Endo Y, Komatsu S, Hirai M, Shimizu N, Suzuki S. Protein kinase C activity and protein phosphorylation in mouse eggs. JOURNAL OF IN VITRO FERTILIZATION AND EMBRYO TRANSFER : IVF 1991; 8:160-6. [PMID: 1919264 DOI: 10.1007/bf01131708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The treatment of mouse eggs with phorbol esters and diacylglycerol inhibits sperm penetration and results in biochemical modification of the zona pellucida. In this report, we have demonstrated the presence of protein kinase C (PKC) activity in mouse eggs as determined by 12-O-tetradecanoyl phorbol-13-acetate (TPA) dependent in vivo and in vitro protein phosphorylation in mouse eggs. When mouse eggs were radiolabeled with [32P]phosphate and treated with TPA, two specific proteins, 70 and 20 kDa, were phosphorylated. The 70-kDa protein was also phosphorylated in vitro by endogenous PKC. In addition, we have shown that exogenous PKC induced the in vitro phosphorylation of 70-, 55-, and 20-kDa proteins in egg extract. The 70-kDa protein was also phosphorylated in vitro after treatment of the cytosol fraction of mouse eggs with TPA, suggesting that this protein might be a specific substrate for PKC and that it is located in the cytosol. These results demonstrate that mouse eggs contain PKC activity and suggest that PKC-catalyzed protein phosphorylation of specific proteins might be involved in the regulation of egg-induced modification of the zona pellucida.
Collapse
Affiliation(s)
- Y Endo
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
48
|
Schwartz DA, Schultz RM. Stimulatory effect of okadaic acid, an inhibitor of protein phosphatases, on nuclear envelope breakdown and protein phosphorylation in mouse oocytes and one-cell embryos. Dev Biol 1991; 145:119-27. [PMID: 1850367 DOI: 10.1016/0012-1606(91)90218-r] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Treatment of one-cell mouse embryos with okadaic acid (OA), which is an inhibitor of protein phosphatases 1 and 2A, induces a concentration-dependent precocious nuclear envelope breakdown (NEBD) of the pronuclei; at 10 microM okadaic acid, NEBD starts to occur after 1 hr and the embryos become committed to NEBD after about 45 min. Correlated with NEBD is the conversion of a protein of Mr 32,000 (p32) to more highly phosphorylated forms. One-cell embryos cultured continuously in OA-containing medium do not cleave, whereas one-cell embryos incubated for 15-60 min prior to transfer to OA-free medium reveal a time-dependent inhibition in their ability to cleave. OA treatment of oocytes that are arrested from resuming spontaneous maturation by either a phosphodiesterase inhibitor or biologically active phorbol diester results in germinal vesicle breakdown and the maturation-associated changes in the pattern of protein phosphorylation, which include the apparent phosphorylation of p32. Results of these experiments implicate protein phosphatases in the G2 to M transition of the cell cycle in both meiotic and mitotic cells.
Collapse
Affiliation(s)
- D A Schwartz
- Department of Biology, University of Pennsylvania, Philadelphia 19104-6018
| | | |
Collapse
|
49
|
Homa ST. Neomycin, an inhibitor of phosphoinositide hydrolysis, inhibits the resumption of bovine oocyte spontaneous meiotic maturation. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1991; 258:95-103. [PMID: 1651367 DOI: 10.1002/jez.1402580111] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The possibility that the intracellular signals generated upon phosphoinositide hydrolysis are involved in regulating bovine oocyte spontaneous meiotic resumption was investigated. Oocytes were mass-harvested and cultured in 2A-BMOC medium supplemented with 0.5% bovine serum albumin in the presence or absence of neomycin (an inhibitor of phosphoinositide hydrolysis) or phorbol myristate acetate (an activator of protein kinase C). The role of intracellular calcium was examined by preloading with BAPTA/AM (a calcium chelator) prior to culture. Meiotic maturation was scored cytogenetically. 1) Neomycin induces an irreversible inhibition of germinal vesicle breakdown which does not exceed 60% and is apparent at concentrations of 5 mM or above. Progression of meiosis past metaphase I is inhibited at concentrations of 2.5 mM or above. The full effect of neomycin is only apparent if it is presented to the oocytes within 3 h of follicular release, although germinal vesicle breakdown is not observed until 9 h culture under control conditions. 2) PMA alone has negligible effect on germinal vesicle breakdown, but it acts synergistically with 2 mM IBMX to inhibit this process. PMA has a dual effect on the progression of meiosis past metaphase I: 1 nM PMA has a stimulatory effect while 1 microM PMA blocks the ability of oocytes to reach anaphase I or beyond. These observations are not found with a non-tumor-promoting phorbol ester. 3) Spontaneous meiotic resumption is not significantly affected in the absence of added exogenous calcium. However, oocytes preloaded with BAPTA/AM exhibit a dose-dependent inhibition of germinal vesicle breakdown, even in the presence of extracellular calcium.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- S T Homa
- Department of Zoology, Arizona State University, Tempe 85287
| |
Collapse
|
50
|
Gavin AC, Tsukitani Y, Schorderet-Slatkine S. Induction of M-phase entry of prophase-blocked mouse oocytes through microinjection of okadaic acid, a specific phosphatase inhibitor. Exp Cell Res 1991; 192:75-81. [PMID: 1701730 DOI: 10.1016/0014-4827(91)90159-r] [Citation(s) in RCA: 65] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We report that a specific inhibitor of types 1 and 2A phosphatases, okadaic acid (OA), induces germinal vesicle break down (GVBD) and chromosome condensation when microinjected into denuded mouse oocytes maintained in prophase block by analogs of cAMP, inhibitors of phosphodiesterase, or a tumor-promoting phorbol ester. GVBD and chromosome condensation are also observed when incompetent oocytes are similarly injected with OA, this effect being dependent on the oocyte diameter. Marked changes in cell shape, cytoskeletal organization, and chromosome condensation with abnormal or abortive spindle formation are associated with such injections. The polar body is not formed. These results led to the conclusions that in mouse oocytes, OA acts distal to both the cAMP-modulated pathway involved in meiotic arrest and the inhibitory action exerted by tumor-promoting phorbol esters.
Collapse
Affiliation(s)
- A C Gavin
- Department of Obstetrics and Gynaecology, Hôpital Cantonal Universitaire, Geneva, Switzerland
| | | | | |
Collapse
|