Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Cells ; 40(11): 871-879, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29145719

ABSTRACT

Levels of maturation-promoting factor (MPF) in oocytes decline after vitrification, and this decline has been suggested as one of the main causes of low developmental competence resulting from cryoinjury. Here, we evaluated MPF activity in vitrified mouse eggs following treatment with caffeine, a known stimulator of MPF activity, and/or the proteasome inhibitor MG132. Collected MII oocytes were vitrified and divided into four groups: untreated, 10 mM caffeine (CA), 10 µM MG132 (MG), and 10 mM caffeine +10 µM MG132 (CA+MG). After warming, the MPF activity of oocytes and their blastocyst formation and implantation rates in the CA, MG, and CA+MG groups were much higher than those in the untreated group. However, the cell numbers in blastocysts did not differ among groups. Analysis of the effectiveness of caffeine and MG132 for improving somatic cell nuclear transfer (SCNT) technology using cryopreserved eggs showed that supplementation did not improve the blastocyst formation rate of cloned mouse eggs. These results suggest that maintaining MPF activity after cryopreservation may have a positive effect on further embryonic development, but is unable to fully overcome cryoinjury. Thus, intrinsic factors governing the developmental potential that diminish during oocyte cryopreservation should be explored.


Subject(s)
Blastocyst/drug effects , Caffeine/pharmacology , Leupeptins/pharmacology , Maturation-Promoting Factor/metabolism , Oocytes/metabolism , Animals , Embryonic Development/drug effects , Female , Fertilization in Vitro , Mesothelin , Mice , Nuclear Transfer Techniques , Oocytes/drug effects , Pregnancy , Up-Regulation , Vitrification
2.
Reprod Sci ; 23(11): 1509-1517, 2016 11.
Article in English | MEDLINE | ID: mdl-27071961

ABSTRACT

Estrogen-related receptor ß (ESRRB), which is a member of the nuclear orphan receptor family, regulates the messenger RNA (mRNA) expression levels of the transcription factors, Oct4 and Nanog, in early embryos and germ cells, thereby maintaining the undifferentiated state and pluripotency of the relevant cells. The present study was designed to determine whether the upregulation of pluripotency-related genes by direct delivery of ESRRB protein may affect on the commitment into inner cell mass (ICM) or the development of vitrified/warmed mouse embryos. Recombinant cell-penetrating peptide (CPP) ESRRB protein was synthesized and then added into a culture medium for cryopreserved mouse embryos. Vitrified/warmed 8-cell embryos were cultured in KSOM with/without 2 µg/mL CPP-ESRRB for 48 hours and then analyzed or transferred to the uteri of foster mothers. The mRNA expression of Oct4 and Nanog was higher in CPP-ESRRB-treated blastocysts compared to the untreated controls. No difference was observed in embryonic development, but ICM:trophectoderm ratio was increased in the CPP-ESRRB-treated group compared to the untreated group, and after embryo transfer, a higher implantation rate was obtained in the CPP-ESRRB-treated group compared to the untreated group. This study shows for the first time that recombinant CPP-ESRRB can be easily integrated into vitrified/warmed mouse embryos and that it increases Oct4 expression (via a pluripotency-related gene pathway), ICM formation, and the further embryonic and full-term development of vitrified/warmed mouse embryos. This CPP-conjugated protein delivery system could therefore be a useful tool for improving assisted reproductive technology.


Subject(s)
Blastocyst/drug effects , Blastocyst/metabolism , Cell-Penetrating Peptides/administration & dosage , Embryonic Development , Estrogens, Conjugated (USP)/administration & dosage , Receptors, Estrogen/administration & dosage , Animals , Cell Count , Culture Media , Embryo Implantation/drug effects , Embryo Transfer , Female , Gene Expression Regulation, Developmental , Male , Mice , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , RNA, Messenger/metabolism , Vitrification
3.
Cell Calcium ; 59(1): 21-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26725171

ABSTRACT

Fertilization in mammals is initiated when a sperm fuses with a mature MII oocyte, also known as egg, and triggers a plethora of finely controlled processes identified as egg activation. The completion of all events of egg activation is driven by and depends on a series of repetitive calcium (Ca(2+)) increases (Ca(2+) oscillations), which rely on Ca(2+) influx from the extracellular media. Ca(2+) channels on the egg plasma membrane (PM) are thought to mediate this influx. The TRP Ca(2+) channel TRPV3 is differentially expressed during oocyte maturation, being most active at the MII stage. Specific stimulation of TRPV3 channels promotes Ca(2+) influx sufficient to induce egg activation and parthenogenesis. Here, we explore the function and distribution dynamics of the TRPV3 channel protein during maturation. Using dsRNA, TrpV3 overexpression, and inhibitors of protein synthesis, we modified the expression levels of the channel and showed that the TRPV3 protein is synthesized and translocated to the PM during maturation. We demonstrated that 2-APB at the concentrations used here to promote Ca(2+) influx in eggs, specifically and reversibly targets TRPV3 channels without blocking IP3R1. Finally, we found that the activity of TRPV3 channels is dependent upon an intact actin cytoskeleton, suggesting an actin-based regulation of its expression and/or function on the PM. Collectively, our results show TRPV3 is a target of 2-APB in eggs, a condition that can be used to induce parthenogenesis. The need of an intact actin cytoskeleton for the function of TRPV3 channels in oocytes is a novel finding and suggests the rearrangements of actin that occur during maturation could regulate both the presence on the PM and/or the function of TRPV3 and of other Ca(2+) channels involved in oocyte maturation and fertilization.


Subject(s)
Boron Compounds/pharmacology , Calcium/metabolism , Oocytes/drug effects , Oocytes/metabolism , TRPV Cation Channels/physiology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Female , Mice , TRPV Cation Channels/antagonists & inhibitors
4.
PLoS One ; 10(12): e0144245, 2015.
Article in English | MEDLINE | ID: mdl-26656301

ABSTRACT

Cisplatin is a first-line chemotherapeutic agent for ovarian cancer that acts by promoting DNA cross links and adduct. However drug resistance and considerable side effects including reproductive toxicity remain a significant challenge. PTEN is well known as a tumor suppressor function which plays a fundamental role in the regulation of the cell cycle, apoptosis and development of cancer. At the same time PTEN has been revealed to be critically important for the maintenance of the primordial follicle pool. In this study, we investigated the role of PTEN/Akt/FOXO3 pathway in cisplatin-induced primordial follicle depletion. Cisplatin induced ovarian failure mouse model was used to evaluate how this pathway involves. In vitro maturation was used for oocyte rescue after cisplatin damage. We found that cisplatin treatment decreased PTEN levels, leading to a subsequent increase in the phosphorylation of key molecules in the pathway. The activation of the PTEN/Akt/FOXO3 pathway cascade increased cytoplasmic translocation of FOXO3a in cisplatin-treated follicles, which in turn increased the pool size of growing follicles, and rapidly depleted the number of dormant follicles. Once activated, the follicles were more prone to apoptosis, and their cumulus cells showed a loss of luteinizing hormone (LH) receptor expression, which leads to failure during final maturation and ovulation. In vitro maturation to rescue oocytes in a cisplatin-treated mouse model resulted in successful maturation and fertilization. This study is the first to show the involvement of the PTEN/Akt/FOXO3 pathway in premature ovarian failure after cisplatin treatment and the possibility of rescue through in vitro maturation.


Subject(s)
Cisplatin/adverse effects , Forkhead Transcription Factors/metabolism , Ovarian Follicle/physiology , Ovarian Reserve/drug effects , PTEN Phosphohydrolase/metabolism , Primary Ovarian Insufficiency/chemically induced , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/physiology , Cisplatin/pharmacology , Female , Fertilization/physiology , Forkhead Box Protein O3 , Mice , Mice, Inbred ICR , Oocytes/physiology , Ovarian Follicle/drug effects , Ovarian Neoplasms/drug therapy , Ovulation/drug effects , Ovulation/physiology , Primary Ovarian Insufficiency/pathology , Receptors, LH/biosynthesis
5.
Clin Exp Reprod Med ; 41(1): 1-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24693491

ABSTRACT

OBJECTIVE: Estrogen related receptor ß (Esrrb) is a member of the orphan nuclear receptors and may regulate the expression of pluripotency-related genes, such as Oct4 and Nanog. Therefore, in the present study, we have developed a method for delivering exogenous ESRRB recombinant protein into embryos by using cell-penetrating peptide (CPP) conjugation and have analyzed their effect on embryonic development. METHODS: Mouse oocytes and embryos were obtained from superovulated mice. The expression of Oct4 mRNA and the cell number of inner cell mass (ICM) in the in vitro-derived and in vivo-derived blastocysts were first analyzed by real time-reverse transcription-polymerase chain reaction and differential staining. Then 8-cell embryos were cultured in KSOM media with or without 2 µg/mL CPP-ESRRB protein for 24 to 48 hours, followed by checking their integration into embryos during in vitro culture by Western blot and immunocytochemistry. RESULTS: Expression of Oct4 and the cell number of ICM were lower in the in vitro-derived blastocysts than in the in vivo-derived ones (p<0.05). In the blastocysts derived from the CPP-ESRRB-treated group, expression of Oct4 was greater than in the non-treated groups (p<0.05). Although no difference in embryonic development was observed between the treated and non-treated groups, the cell number of ICM was greater in the CPP-ESRRB-treated group. CONCLUSION: Treatment of CPP-ESRRB during cultivation could increase embryos' expression of Oct4 and the formation rate of the ICM in the blastocyst. Additionally, an exogenous delivery system of CPP-conjugated protein would be a useful tool for improving embryo culture systems.

6.
Reprod Sci ; 21(7): 859-868, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24401473

ABSTRACT

The aim of this study was to evaluate the effectiveness of improving angiogenesis at graft sites on the survival of follicles in transplanted ovarian tissue. Matrigel containing 5 × 105 of cord blood-derived endothelial progenitor cells (EPCs) or 200 ng of mouse vascular endothelial growth factor (VEGF) was injected subcutaneously into BALB/c-Nu mice. After 1 week, vitrified/warmed ovaries from female B6D2F1 mice were subcutaneously transplanted into the injection sites. After 1, 2, and 4 weeks posttransplantation, the ovaries were recovered and subjected to histological analysis. Oocytes were collected from the transplanted ovaries, and their fertilization, embryonic development, and delivery were also observed. Vitrified/warmed ovaries transplanted into EPC- or VEGF-treated sites developed more blood vessels and showed better follicle survival than those transplanted into sham-injected sites. Normal embryonic development and consequent live births were obtained using oocytes recovered from cryopreserved/transplanted ovaries. Treatment with EPCs or VEGF could prevent the ischemic damage during the early revascularization stage of ovarian transplantation.

7.
Toxicol Appl Pharmacol ; 265(1): 61-72, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23022515

ABSTRACT

Fluoxetine, a selective serotonin reuptake inhibitor, regulates a variety of physiological processes, such as cell proliferation and apoptosis, in mammalian cells. Little is known about the role of fluoxetine in early embryonic development. This study was undertaken to investigate the effect of fluoxetine during mouse early embryonic development. Late two-cell stage embryos (2-cells) were cultured in the presence of various concentrations of fluoxetine (1 to 50µM) for different durations. When late 2-cells were incubated with 5µM fluoxetine for 6h, the percentage that developed into blastocysts increased compared to the control value. However, late 2-cells exposed to fluoxetine (5µM) over 24h showed a reduction in blastocyst formation. The addition of fluoxetine (5µM) together with KN93 or KN62 (calcium/calmodulin-dependent protein kinase II (CaMKII) inhibitors) failed to increase blastocyst formation. Fluoxetine treatment inhibited TREK-1 and TREK-2, members of the two-pore domain K(+) channel family expressed in mouse embryos, activities, indicating that fluoxetine-induced membrane depolarization in late 2-cells might have resulted from TREK inhibition. In addition, long-term exposure to fluoxetine altered the TREK mRNA expression levels. Furthermore, injection of siRNA targeting TREKs significantly decreased blastocyst formation by ~30% compared to injection of scrambled siRNA. Long-term exposure of fluoxetine had no effect on blastocyst formation of TREK deficient embryos. These results indicate that low-dose and short-term exposures of late 2-cells to fluoxetine probably increase blastocyst formation through activation of CaMKII-dependent signal transduction pathways, whereas long-term exposure decreases mouse early embryonic development through inhibition of TREK channel gating.


Subject(s)
Embryonic Development/drug effects , Fluoxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Blastocyst/drug effects , Blotting, Western , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chromosomes/drug effects , Enzyme Activation/drug effects , Female , Membrane Potentials/drug effects , Mice , Oxygen Consumption/drug effects , Patch-Clamp Techniques , Polymerase Chain Reaction , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Pregnancy , RNA, Small Interfering/genetics , Signal Transduction/drug effects
8.
Reproduction ; 143(5): 625-36, 2012 May.
Article in English | MEDLINE | ID: mdl-22419831

ABSTRACT

Numerous studies have suggested that K(+) channels regulate a wide range of physiological processes in mammalian cells. However, little is known about the specific function of K(+) channels in germ cells. In this study, mouse zygotes were cultured in a medium containing K(+) channel blockers to identify the functional role of K(+) channels in mouse embryonic development. Voltage-dependent K(+) channel blockers, such as tetraethylammonium and BaCl(2), had no effect on embryonic development to the blastocyst stage, whereas K(2P) channel blockers, such as quinine, selective serotonin reuptake inhibitors (fluoxetine, paroxetine, and citalopram), gadolinium trichloride, anandamide, ruthenium red, and zinc chloride, significantly decreased blastocyst formation (P<0.05). RT-PCR data showed that members of the K(2P) channel family, specifically KCNK2, KCNK10, KCNK4, KCNK3, and KCNK9, were expressed in mouse oocytes and embryos. In addition, their mRNA expression levels, except Kcnk3, were up-regulated by above ninefold in morula-stage embryos compared with 2-cell stage embryos (2-cells). Immunocytochemical data showed that KCNK2, KCNK10, KCNK4, KCNK3, and KCNK9 channel proteins were expressed in the membrane of oocytes, 2-cells, and blastocysts. Each siRNA injection targeted at Kcnk2, Kcnk10, Kcnk4, Kcnk3, and Kcnk9 significantly decreased blastocyst formation by ~38% compared with scrambled siRNA injection (P<0.05). The blockade of K(2P) channels acidified the intracellular pH and depolarized the membrane potential. These results suggest that K(2P) channels could improve mouse embryonic development through the modulation of gating by activators.


Subject(s)
Blastocyst/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Zygote/metabolism , Animals , Blastocyst/drug effects , Embryo Culture Techniques , Embryonic Development , Gene Expression Regulation, Developmental , Hydrogen-Ion Concentration , Immunohistochemistry , Ion Channel Gating , Membrane Potentials , Mice , Mice, Inbred ICR , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , RNA Interference , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Zygote/drug effects
9.
Hum Reprod ; 27(6): 1768-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22456923

ABSTRACT

BACKGROUND: Oocyte activation is a crucial step that comprises the release of the oocyte from meiotic arrest, pronuclear formation and subsequent embryo development. Oocytes are activated by repetitive increases in the intracellular concentration of free Ca(2+), [Ca(2+)](i) oscillations, which are triggered during fertilization by the introduction of the sperm-specific phospholipase C zeta 1 (PLCZ1). Recent studies have shown that sperm from patients lacking expression of PLCZ1 or expressing mutant forms of PLCZ1 fail to induce [Ca(2+)](i) oscillations or oocyte activation. We first purified recombinant human PLCZ1 (hPLCZ1) protein and evaluated its [Ca(2+)](i) oscillation activity in mouse and human oocytes with the view to investigate its application in the clinic for assisted oocytes activation in lieu of chemical agents. METHODS: Recombinant hPLCZ1 was synthesized using the Escherichia coli system, and subjected to immunoblot analysis with anti-PLCZ1 and anti-His tag antibodies. [Ca(2+)](i) oscillations by microinjection of recombinant hPLCZ1 into mouse or human oocytes were examined by [Ca(2+)](i) monitoring with Fluo 4. Ploidy of the oocytes with recombinant hPLCZ1 injection was confirmed with fluorescence in situ hybridization. RESULTS: A band of 68 kDa on recombinant protein was detected with both antibodies. Injection of recombinant hPLCZ1 induced [Ca(2+)](i) oscillations in a dose-dependent manner in both mouse and human oocytes. These oscillations, which closely resembled those initiated by the sperm upon fertilization, triggered activation and cleavage in oocytes of both species, although further development of the mice embryos was low. U73122, a PLC inhibitor, blocked the ability of hPLCZ1 to initiate oscillations. Microinjection of recombinant hPLCZ1 into ICSI-failed human oocytes rescued fertilization failure in five of eight attempts. CONCLUSIONS: Repeated [Ca(2+)](i) oscillations and oocyte activation were induced in mouse and human oocytes by microinjection of recombinant hPLCZ1 synthesized in E. Coli. Injection of recombinant protein could thus provide a biological solution for inducing artificial activation of oocytes.


Subject(s)
Calcium Signaling/drug effects , Oocytes/drug effects , Oocytes/physiology , Phosphoinositide Phospholipase C/pharmacology , Recombinant Proteins/pharmacology , Adult , Animals , Calcium/metabolism , Female , Fertilization in Vitro , Humans , Male , Mice
10.
J Cell Physiol ; 227(2): 705-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21465476

ABSTRACT

At the time of fertilization, an increase in the intracellular Ca(2+) concentration ([Ca(2+)](i)) underlies egg activation and initiation of development in all species studied to date. The inositol 1,4,5-trisphosphate receptor (IP(3)R1), which is mostly located in the endoplasmic reticulum (ER) mediates the majority of this Ca(2+) release. The sensitivity of IP(3)R1, that is, its Ca(2+) releasing capability, is increased during oocyte maturation so that the optimum [Ca(2+)](i) response concurs with fertilization, which in mammals occurs at metaphase of second meiosis. Multiple IP(3)R1 modifications affect its sensitivity, including phosphorylation, sub-cellular localization, and ER Ca(2+) concentration ([Ca(2+)](ER)). Here, we evaluated using mouse oocytes how each of these factors affected IP(3)R1 sensitivity. The capacity for IP(3)-induced Ca(2+) release markedly increased at the germinal vesicle breakdown stage, although oocytes only acquire the ability to initiate fertilization-like oscillations at later stages of maturation. The increase in IP(3)R1 sensitivity was underpinned by an increase in [Ca(2+)](ER) and receptor phosphorylation(s) but not by changes in IP(3)R1 cellular distribution, as inhibition of the former factors reduced Ca(2+) release, whereas inhibition of the latter had no impact. Therefore, the results suggest that the regulation of [Ca(2+)](ER) and IP(3)R1 phosphorylation during maturation enhance IP(3)R1 sensitivity rendering oocytes competent to initiate oscillations at the expected time of fertilization. The temporal discrepancy between the initiation of changes in IP(3)R1 sensitivity and acquisition of mature oscillatory capacity suggest that other mechanisms that regulate Ca(2+) homeostasis also shape the pattern of oscillations in mammalian eggs.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/metabolism , Oocytes/cytology , Oocytes/physiology , Animals , Calcium Signaling/physiology , Cyclin-Dependent Kinase Inhibitor Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Female , Gene Expression Regulation/physiology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Phosphorylation , Protein Transport
11.
PLoS One ; 6(8): e23304, 2011.
Article in English | MEDLINE | ID: mdl-21850267

ABSTRACT

Previously, we found that the growth arrest-specific gene 6 (Gas6) is more highly expressed in germinal vesicle (GV) oocytes than in metaphase II (MII) oocytes using annealing control primer (ACP)-PCR technology. The current study was undertaken to investigate the role of Gas6 in oocyte maturation and fertilization using RNA interference (RNAi). Interestingly, despite the specific and marked decrease in Gas6 mRNA and protein expression in GVs after Gas6 RNAi, nuclear maturation including spindle structures and chromosome segregation was not affected. The only discernible effect induced by Gas6 RNAi was a change in maturation promoting factor (MPF) activity. After parthenogenetic activation, Gas6 RNAi-treated oocytes at the MII stage had not developed further and arrested at MII (90.0%). After stimulation with Sr(2+), Gas6-silenced MII oocytes had markedly reduced Ca(2+) oscillation and exhibited no exocytosis of cortical granules. In these oocytes, sperm penetration occurred during fertilization but not pronucleus (PN) formation. By roscovitine and colcemid treatment, we found that the Gas6 knockdown affected cytoplasmic maturation directly, independent to the changed MPF activity. These results strongly suggest that 1) the Gas6 signaling itself is important to the cytoplasmic maturation, but not nuclear maturation, and 2) the decreased Gas6 expression and decreased MPF activity separately or mutually influence sperm head decondensation and PN formation.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Maturation-Promoting Factor/metabolism , Animals , Blotting, Western , Cells, Cultured , Chromosomes/metabolism , Embryo, Mammalian/metabolism , Female , Fertilization/genetics , Fertilization/physiology , Intercellular Signaling Peptides and Proteins/genetics , Male , Maturation-Promoting Factor/genetics , Mesothelin , Mice , Oocytes/metabolism , Pregnancy , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
12.
Clin Exp Pharmacol Physiol ; 38(7): 435-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545631

ABSTRACT

1. In mice, acetylcholine (ACh) plays an important role in oocyte activation and embryonic development. However, the role of ACh in mouse oocyte maturation has not been investigated. 2. In the present study, the effects of 100 µmol/L and 1 mmol/L ACh on maturation processes of murine germinal vesicle (GV) intact oocytes (GV oocytes) exposed to 10 and 100 µmol/L 3-isobutyl-1-methylxanthine (IBMX), an inhibitor of cyclic nucleotide phosphodiesterase, were evaluated morphologically and immunologically. It has been shown that IBMX inhibits the resumption of meiosis by preventing cAMP breakdown. 3. In the present study, at the start of in vitro culture 100% of oocytes were at the GV stage. After 18 h culture, 95 ± 3, 0 and 85.8 ± 10.2% of oocytes had passed the GV stage in the control, IBMX and IBMX + ACh groups, respectively. The IBMX-induced inhibition of the maturation process was significantly attenuated by approximately 90% by ACh in groups treated with 10 µmol/L IBMX + 100 µmol/L ACh and 100 µmol/L IBMX + 1 mmol/L ACh. Although cAMP levels were high in oocytes treated with 100 µmol/L IBMX, levels were reduced in groups treated simultaneously with 100 µmol/L ACh. Furthermore, compared with mature oocytes, ACh-treated GV oocytes exhibited significantly lower (by approximately 2.3-fold) or absent Ca(2+) peaks. 4. The results of the present study indicate that maturation of GV oocytes, arrested by IBMX treatment, is resumed following ACh treatment and that this effect is due to downregulation of cAMP rather than changes in intracellular Ca(2+) levels.


Subject(s)
Acetylcholine/pharmacology , Acetylcholine/physiology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Oocytes/drug effects , Oocytes/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cells, Cultured , Down-Regulation , Female , Meiosis/drug effects , Mice , Mice, Inbred ICR , Oocytes/cytology , Oocytes/metabolism
13.
Pflugers Arch ; 461(5): 515-26, 2011 May.
Article in English | MEDLINE | ID: mdl-21431324

ABSTRACT

Cryopreservation of mature eggs is a useful technique that can be applied in assisted reproductive technology. However, the method has some limitations, such as cryodamage induced by biophysical modifications during the cryopreservation process. To assess these biophysical damage, we analyzed the relationship between intracellular calcium ([Ca2+]i) oscillatory activity via type 1 inositol 1,4,5-trisphosphate receptor (IP(3)R1) distribution after vitrification and efficiency of cryopreservation according to cryoprotectant (CPA) composition. In immunostaining, results of IP(3)R1 with eggs after the vitrification performed using ethylene glycol (EG) alone or EG + dimethylsulfoxide (DMSO) as CPAs, CPA-treated, and fresh eggs displayed a homogeneous IP(3)R1 distribution which is spread uniformly throughout cytoplasm with clusters on the cortex. However, after vitrification and warming process, more than 60% of eggs displayed a heterogeneous distribution which is non-uniform distribution with patches and disconnection of IP(3)R1. In 90-min incubation for recovery from cryodamage, eggs from the EG + DMSO group recovered from with a heterogeneous IP(3)R1 distribution to the homogeneous distribution, but not in EG alone group. In ICSI experiments, vitrified eggs in the EG-alone group presented significantly low blastocyst formation compared to those of the fresh and EG + DMSO groups. These results suggest that the vitrification process influences IP(3)R1 distribution, and subsequently, [Ca2+]i oscillatory activity and embryonic development. Accordingly, we propose that IP(3)R1 distribution and [Ca2+]i oscillatory activity are correlated with egg quality and developmental potential after vitrification, and may thus be applied as an effective indicator to evaluate the efficiency of oocyte cryopreservation methods.


Subject(s)
Cryoprotective Agents/pharmacology , Embryonic Development/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ovum/drug effects , Animals , Calcium Signaling/drug effects , Dimethyl Sulfoxide/pharmacology , Ethylene Glycol/pharmacology , Female , Fertilization in Vitro , Male , Mice , Sperm Injections, Intracytoplasmic , Vitrification
14.
J Cell Physiol ; 222(1): 238-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19798695

ABSTRACT

The initiation of normal embryo development depends on the completion of all events of egg activation. In all species to date, egg activation requires an increase(s) in the intracellular concentration of calcium ([Ca(2+)](i)), which is almost entirely mediated by inositol 1,4,5-trisphosphate receptor 1 (IP(3)R1). In mammalian eggs, fertilization-induced [Ca(2+)](i) responses exhibit a periodic pattern that are called [Ca(2+)](i) oscillations. These [Ca(2+)](i) oscillations are robust at the beginning of fertilization, which occurs at the second metaphase of meiosis, but wane as zygotes approach the pronuclear stage, time after which in the mouse oscillations cease altogether. Underlying this change in frequency are cellular and biochemical changes associated with egg activation, including degradation of IP(3)R1, progression through the cell cycle, and reorganization of intracellular organelles. In this study, we investigated the system requirements for IP(3)R1 degradation and examined the impact of the IP(3)R1 levels on the pattern of [Ca(2+)](i) oscillations. Using microinjection of IP(3) and of its analogs and conditions that prevent the development of [Ca(2+)](i) oscillations, we show that IP(3)R1 degradation requires uniform and persistently elevated levels of IP(3). We also established that progressive degradation of the IP(3)R1 results in [Ca(2+)](i) oscillations with diminished periodicity while a near complete depletion of IP(3)R1s precludes the initiation of [Ca(2+)](i) oscillations. These results provide insights into the mechanism involved in the generation of [Ca(2+)](i) oscillations in mouse eggs.


Subject(s)
Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ovum/metabolism , Protein Processing, Post-Translational , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Calcium Signaling/drug effects , Down-Regulation/drug effects , Female , Fertilization/drug effects , Humans , Injections , Inositol 1,4,5-Trisphosphate/pharmacology , Mice , Ovum/drug effects , Protein Processing, Post-Translational/drug effects , Time Factors
15.
J Cell Sci ; 122(Pt 15): 2741-9, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19596796

ABSTRACT

One of the most important processes in fertilization is the fusion of egg and sperm; however, the molecular mechanisms involved in this process are not well understood. So far, using genetic approaches, only two proteins have been demonstrated to be necessary for this process: Izumo in sperm and CD9 in the egg. Here we demonstrate that sperm produced by Tssk6 (Sstk)-null mice present defects that prevent the successful fertilization of eggs in vitro and the fusion to zona-pellucida-free eggs. Tssk6 is a member of the testis-specific serine kinase family of proteins and is expressed postmeiotically in male germ cells. In order for fusion to occur, during the process known as acrosome reaction Izumo needs to relocate from the anterior head to other regions, including the postacrosomal compartment. Tssk6-null sperm fails to relocate Izumo during the acrosome reaction. Agents that interfere with actin dynamics blocked the acrosome-reaction-associated translocation of Izumo that is required for fusion in wild-type sperm. Additionally, actin polymerization was compromised in Tssk6-null sperm. Taken together, our results indicate that Tssk6 is involved in sperm-egg fusion through the regulation of actin polymerization and changes in Izumo localization.


Subject(s)
Immunoglobulins/physiology , Membrane Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Sperm-Ovum Interactions/physiology , Spermatozoa/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Fusion , Female , Fertilization in Vitro , Immunoblotting , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatozoa/cytology
16.
Pflugers Arch ; 458(6): 1125-36, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19484474

ABSTRACT

Acetylcholine (ACh) causes early activation events in mouse oocytes, but little is known about its precise role in the early embryonic development of mice. We aimed to determine whether and how ACh is capable of rescuing two-cell block in an in vitro culture system. ACh evoked different transient Ca(2+) patterns showing a higher Ca(2+) peak in the two-cell stage embryos (two-cells) than observed in mature oocytes. In early two-cells subjected to an in vitro two-cell block, xestospongin C (Xes-C), an IP3 receptor antagonist, significantly decreased the level of the ACh-induced Ca(2+) increase. The reduction in the ACh-induced Ca(2+) increase by Xes-C in late two-cells was lower than that in early two-cells. Furthermore, KN62 and KN93, both CaMKII inhibitors, were found to reduce the magnitude of the ACh-induced Ca(2+) increase in early two-cells. The addition of ACh to the culture medium showed an ability to rescue in vitro two-cell block. However, the addition of ACh together with both Xes-C and CaMKII inhibitors or with either inhibitor separately had no effect on the rescue of two-cell block. Long-term exposure of late two-cells to ACh decreased morula and early blastocyst development and ACh had a differential effect on early and late two-cells. These results indicate that ACh likely rescues the in vitro two-cell block through activation of IP3R- and/or CaMKII-dependent signal transduction pathways.


Subject(s)
Acetylcholine/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Inositol 1,4,5-Trisphosphate Receptors/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Benzylamines/pharmacology , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Embryonic Development/drug effects , Female , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Macrocyclic Compounds/pharmacology , Male , Mice , Mice, Inbred ICR , Oxazoles/pharmacology , Sulfonamides/pharmacology
17.
Dev Cell ; 16(6): 856-66, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531356

ABSTRACT

Xenopus oocyte death is partly controlled by the apoptotic initiator caspase-2 (C2). We reported previously that oocyte nutrient depletion activates C2 upstream of mitochondrial cytochrome c release. Conversely, nutrient-replete oocytes inhibit C2 via S135 phosphorylation catalyzed by calcium/calmodulin-dependent protein kinase II. We now show that C2 phosphorylated at S135 binds 14-3-3zeta, thus preventing C2 dephosphorylation. Moreover, we determined that S135 dephosphorylation is catalyzed by protein phosphatase-1 (PP1), which directly binds C2. Although C2 dephosphorylation is responsive to metabolism, neither PP1 activity nor binding is metabolically regulated. Rather, release of 14-3-3zeta from C2 is controlled by metabolism and allows for C2 dephosphorylation. Accordingly, a C2 mutant unable to bind 14-3-3zeta is highly susceptible to dephosphorylation. Although this mechanism was initially established in Xenopus, we now demonstrate similar control of murine C2 by phosphorylation and 14-3-3 binding in mouse eggs. These findings provide an unexpected evolutionary link between 14-3-3 and metabolism in oocyte death.


Subject(s)
14-3-3 Proteins/metabolism , Apoptosis , Caspase 2/metabolism , Oocytes/cytology , Oocytes/enzymology , Protein Phosphatase 1/metabolism , Animals , Enzyme Activation , Female , Mice , Phosphorylation , Protein Binding , Xenopus
18.
Reproduction ; 137(3): 427-37, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19074500

ABSTRACT

The production of cloned animals by the transfer of a differentiated somatic cell into an enucleated oocyte circumvents fertilization. During fertilization, the sperm delivers a sperm-specific phospholipase C (PLCZ) that is responsible for triggering Ca(2)(+) oscillations and oocyte activation. During bovine somatic cell nuclear transfer (SCNT), oocyte activation is artificially achieved by combined chemical treatments that induce a monotonic rise in intracellular Ca(2)(+) and inhibit either phosphorylation or protein synthesis. In this study, we tested the hypothesis that activation of bovine nuclear transfer embryos by PLCZ improves nuclear reprogramming. Injection of PLCZ cRNA into bovine SCNT units induced Ca(2)(+) oscillations similar to those observed after fertilization and supported high rates of blastocyst development similar to that seen in embryos produced by IVF. Furthermore, gene expression analysis at the eight-cell and blastocyst stages revealed a similar expression pattern for a number of genes in both groups of embryos. Lastly, levels of trimethylated lysine 27 at histone H3 in blastocysts were higher in bovine nuclear transfer embryos activated using cycloheximide and 6-dimethylaminopurine (DMAP) than in those activated using PLCZ or derived from IVF. These results demonstrate that exogenous PLCZ can be used to activate bovine SCNT-derived embryos and support the hypothesis that a fertilization-like activation response can enhance some aspects of nuclear reprogramming.


Subject(s)
Blastocyst/physiology , Nuclear Transfer Techniques , RNA, Complementary/administration & dosage , Type C Phospholipases/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Calcium/metabolism , Cattle , Cells, Cultured , Cycloheximide/pharmacology , Embryonic Development/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Histones/metabolism , Injections , Methylation
19.
Reprod Fertil Dev ; 20(8): 875-83, 2008.
Article in English | MEDLINE | ID: mdl-19007551

ABSTRACT

Methods presently used to activate mare oocytes for assisted reproduction technologies provide low rates of advanced embryonic development. Because phospholipase Czeta (PLCzeta) is the postulated sperm-borne factor responsible for oocyte activation at fertilisation, the aim of the present study was to investigate the pattern of [Ca(2+)](i) oscillations and developmental rates achieved by microinjection of three concentrations of mouse PLCzeta complementary (c) RNA (1, 0.5 or 0.25 microg microL(-1)) into mare oocytes. The frequency of [Ca(2+)](i) oscillations was no different (P > 0.05) after injection of 1, 0.5 or 0.25 microg microL(-1) PLCzeta cRNA (41.1 +/- 5.3, 47 +/- 4.0 and 55.4 +/- 9.0, respectively). However, [Ca(2+)](i) oscillations persisted longest (P < 0.05) for oocytes injected with 0.5 microg microL(-1) PLCzeta cRNA (570.7 +/- 64.2 min). There was no significant difference in cleavage rates after injection of the three concentrations of PLCzeta (P > 0.05; range 97-100%), but the proportion of oocytes reaching advanced stages of embryonic development (>64 nuclei) was significantly lower for oocytes injected with 0.25 microg microL(-1) PLCzeta cRNA (3%) than for those injected with 1 microg microL(-1) PLCzeta cRNA (15%). Based on these results, microinjection of PLCzeta may prove an effective and consistent method for the parthenogenetic activation of mare oocytes for nuclear transfer and provides a physiologically relevant tool with which to study fertilisation-dependent [Ca(2+)](i) signalling in this species.


Subject(s)
Calcium Signaling/drug effects , Embryonic Development/drug effects , Horses/physiology , Oocytes/drug effects , Phosphoinositide Phospholipase C/genetics , RNA, Complementary/pharmacology , Reproductive Techniques, Assisted/veterinary , Animals , Calcium Signaling/physiology , Dose-Response Relationship, Drug , Embryonic Development/physiology , Female , Mice , Microinjections , Oocytes/physiology , Phosphoinositide Phospholipase C/pharmacology , RNA, Complementary/administration & dosage
20.
J Clin Invest ; 118(11): 3671-81, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18924610

ABSTRACT

Egg activation, which is the first step in the initiation of embryo development, involves both completion of meiosis and progression into mitotic cycles. In mammals, the fertilizing sperm delivers the activating signal, which consists of oscillations in free cytosolic Ca(2+) concentration ([Ca(2+)](i)). Intracytoplasmic sperm injection (ICSI) is a technique that in vitro fertilization clinics use to treat a myriad of male factor infertility cases. Importantly, some patients who repeatedly fail ICSI also fail to induce egg activation and are, therefore, sterile. Here, we have found that sperm from patients who repeatedly failed ICSI were unable to induce [Ca(2+)](i) oscillations in mouse eggs. We have also shown that PLC, zeta 1 (PLCZ1), the sperm protein thought to induce [Ca(2+)](i) oscillations, was localized to the equatorial region of wild-type sperm heads but was undetectable in sperm from patients who had failed ICSI. The absence of PLCZ1 in these patients was further confirmed by Western blot, although genomic sequencing failed to reveal conclusive PLCZ1 mutations. Using mouse eggs, we reproduced the failure of sperm from these patients to induce egg activation and rescued it by injection of mouse Plcz1 mRNA. Together, our results indicate that the inability of human sperm to initiate [Ca(2+)](i) oscillations leads to failure of egg activation and sterility and that abnormal PLCZ1 expression underlies this functional defect.


Subject(s)
Calcium/metabolism , Embryonic Development/genetics , Phosphoinositide Phospholipase C/genetics , Spermatozoa/physiology , Humans , Male , Phosphoinositide Phospholipase C/metabolism , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...