Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 44
1.
Cells ; 13(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38727294

Information on long-term effects of postovulatory oocyte aging (POA) on offspring is limited. Whether POA affects offspring by causing oxidative stress (OS) and mitochondrial damage is unknown. Here, in vivo-aged (IVA) mouse oocytes were collected 9 h after ovulation, while in vitro-aged (ITA) oocytes were obtained by culturing freshly ovulated oocytes for 9 h in media with low, moderate, or high antioxidant potential. Oocytes were fertilized in vitro and blastocysts transferred to produce F1 offspring. F1 mice were mated with naturally bred mice to generate F2 offspring. Both IVA and the ITA groups in low antioxidant medium showed significantly increased anxiety-like behavior and impaired spatial and fear learning/memory and hippocampal expression of anxiolytic and learning/memory-beneficial genes in both male and female F1 offspring. Furthermore, the aging in both groups increased OS and impaired mitochondrial function in oocytes, blastocysts, and hippocampus of F1 offspring; however, it did not affect the behavior of F2 offspring. It is concluded that POA caused OS and damaged mitochondria in aged oocytes, leading to defects in anxiety-like behavior and learning/memory of F1 offspring. Thus, POA is a crucial factor that causes psychological problems in offspring, and antioxidant measures may be taken to ameliorate the detrimental effects of POA on offspring.


Behavior, Animal , Mitochondria , Oocytes , Oxidative Stress , Animals , Oocytes/metabolism , Mitochondria/metabolism , Female , Mice , Male , Ovulation , Anxiety/metabolism , Anxiety/pathology , Antioxidants/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Blastocyst/metabolism , Cellular Senescence , Memory
2.
Theriogenology ; 220: 84-95, 2024 May.
Article En | MEDLINE | ID: mdl-38490113

Understanding the mechanisms for oocyte maturation and optimizing the protocols for in vitro maturation (IVM) are greatly important for improving developmental potential of IVM oocytes. The miRNAs expressed in cumulus cells (CCs) play important roles in oocyte maturation and may be used as markers for selection of competent oocytes/embryos. Although a recent study from our group identified several new CCs-expressed miRNAs that regulate cumulus expansion (CE) and CC apoptosis (CCA) in mouse oocytes, validation of these findings and further investigation of mechanisms of action in other model species was essential before wider applications. By using both in vitro and in vivo pig oocyte models with significant differences in CE, CCA and developmental potential, the present study validated that miR-149 and miR-31 improved CE and developmental potential while suppressing CCA of pig oocytes. We demonstrated that miR-149 and miR-31 targeted SMAD family member 6 (SMAD6) and transforming growth factor ß2 (TGFB2), respectively, in the transforming growth factor-ß (TGF-ß) signaling. Furthermore, both miR-149 and miR-31 increased CE and decreased CCA via activating SMAD family member 2 (SMAD2) and increasing the expression of SMAD2 and SMAD family member 4. In conclusion, the present results show that miR-149 and miR-31 improved CE and developmental potential while suppressing CCA of pig oocytes by activating the TGF-ß signaling, suggesting that they might be used as markers for pig oocyte quality.


Cumulus Cells , In Vitro Oocyte Maturation Techniques , MicroRNAs , Oocytes , Animals , Female , Cumulus Cells/physiology , In Vitro Oocyte Maturation Techniques/veterinary , In Vitro Oocyte Maturation Techniques/methods , MicroRNAs/genetics , MicroRNAs/metabolism , Oocytes/physiology , Swine , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/metabolism
3.
Biol Reprod ; 110(5): 895-907, 2024 May 09.
Article En | MEDLINE | ID: mdl-38267362

It is known that the oocyte has a limited capacity to acquire and metabolize glucose, and it must rely on cumulus cells (CCs) to take up glucose and produce pyruvate for use to produce ATP through oxidative phosphorylation. We therefore propose that miRNAs might regulate glucose metabolism (GM) in CCs and might be used as markers for oocyte quality assessment. Here, mouse CC models with impaired glycolysis or pentose phosphate pathway (PPP) were established, and miRNAs targeting the key enzymes in glycolysis/PPP were predicted using the miRNA target prediction databases. Expression of the predicted miRNAs was compared between CCs with normal and impaired glycolysis/PPP to identify candidate miRNAs. Function of the candidate miRNAs was validated by transfecting CCs or cumulus-oocyte-complexes (COCs) with miRNA inhibitors and observing effects on glucose metabolites of CCs and on competence of oocytes. The results validated that miR-23b-3p, let-7b-5p, 34b-5p and 145a-5p inhibited glycolysis, and miR-24-3p, 3078-3p,183-5p and 7001-5p inhibited PPP of CCs. Our observation using a more physiologically relevant model (intact cultured COCs) further validated the four glycolysis-targeting miRNAs we identified. Furthermore, miR-let-7b-5p, 34b-5p and 145a-5p may also inhibit PPP, as they decreased the production of glucose-6-phosphate. In conclusion, miRNAs play critical roles in GM of CCs and may be used as markers for oocyte quality assessment. Summary sentence:  We identified and validated eight new miRNAs that inhibit glycolysis and/or pentose phosphate pathways in cumulus cells (CCs) suggesting that miRNAs play critical roles in glucose metabolism of CCs and may be used for oocyte quality markers.


Cumulus Cells , Glucose , Glycolysis , MicroRNAs , Animals , Cumulus Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Mice , Glucose/metabolism , Female , Glycolysis/physiology , Pentose Phosphate Pathway , Oocytes/metabolism
4.
J Reprod Dev ; 69(4): 185-191, 2023 Aug 11.
Article En | MEDLINE | ID: mdl-37245986

The mechanisms underlying postovulatory oocyte aging (POA) remain largely unknown. The expression of the calcium-sensing receptor (CaSR) in mouse oocytes and its role in POA need to be explored. Our objective was to observe CaSR expression and its role in the susceptibility to activating stimuli (STAS) in POA mouse oocytes. The results showed that, although none of the newly ovulated oocytes were activated, 40% and 94% of the oocytes recovered 19 and 25 h after human chorionic gonadotropin (hCG) injection were activated, respectively, after ethanol treatment. The level of the CaSR functional dimer protein in oocytes increased significantly from 13 to 25 h post hCG. Thus, the CaSR functional dimer level was positively correlated with the STAS of POA oocytes. Aging in vitro with a CaSR antagonist suppressed the elevation of STAS, and cytoplasmic calcium in oocytes recovered 19 h post hCG, whereas aging with a CaSR agonist increased STAS, and cytoplasmic calcium of oocytes recovered 13 h post hCG. Furthermore, the CaSR was more important than the Na-Ca2+ exchanger in regulating oocyte STAS, and T- and L-type calcium channels were inactive in aging oocytes. We conclude that the CaSR is involved in regulating STAS in POA mouse oocytes, and that it is more important than the other calcium channels tested in this connection.


Calcium , Receptors, Calcium-Sensing , Humans , Animals , Mice , Oocytes , Ovulation , Aging , Polymers
5.
Cells ; 12(3)2023 01 31.
Article En | MEDLINE | ID: mdl-36766799

Although studies indicate that female stress-increased secretion of glucocorticoids impairs oocyte competence and embryo development, by inducing apoptosis of ovarian and oviductal cells, respectively, the mechanisms by which glucocorticoids induce apoptosis of ovarian and oviductal cells are largely unclear. Tissue plasminogen activator (tPA) has been involved in apoptosis of different cell types. However, while some studies indicate that tPA is proapoptotic, others demonstrate its antiapoptotic effects. This study has explored the role and action mechanisms of tPA in corticosterone-induced apoptosis of mouse mural granulosa cells (MGCs) and oviductal epithelial cells (OECs). The results demonstrate that culture with corticosterone significantly increased apoptosis, while decreasing levels of tPA (Plat) mRNA and tPA protein in both MGCs and OECs. Culture with tPA ameliorated corticosterone-induced apoptosis of MGCs and OECs. Furthermore, while tPA protected MGCs from corticosterone-induced apoptosis by interacting with low-density lipoprotein receptor-related protein 1 (LRP1), it protected OECs from the apoptosis by acting on Annexin 2 (ANXA2). In conclusion, tPA is antiapoptotic in both MGCs and OECs, and it protects MGCs and OECs from corticosterone-induced apoptosis by interacting with LRP1 and ANXA2, respectively, suggesting that tPA may use different receptors to inhibit apoptosis in different cell types.


Corticosterone , Glucocorticoids , Animals , Female , Mice , Apoptosis , Corticosterone/pharmacology , Epithelial Cells/metabolism , Glucocorticoids/pharmacology , Tissue Plasminogen Activator/metabolism
6.
Aging (Albany NY) ; 14(22): 9186-9199, 2022 11 26.
Article En | MEDLINE | ID: mdl-36441534

Studies suggest that psychological stress on women can impair their reproduction and that postovulatory oocyte aging (POA) might increase the risk of early pregnancy loss and affect offspring's reproductive fitness and longevity. However, whether psychological stress during oocyte development would facilitate POA is unknown but worth exploring to understand the mechanisms by which psychological stress and POA damage oocytes. This study observed effects of female restraint stress during oocyte development (FRSOD) on oocyte resistance to POA. Female mice were restrained for 48 h before superovulation, and they were sacrificed at different intervals after ovulation to recover aging oocytes for analyzing their early and late aged characteristics. The effects of FRSOD on aging oocytes included: (1) increasing their susceptibility to activation stimulus with elevated cytoplasmic calcium; (2) impairing their developmental potential with downregulated expression of development-beneficial genes; (3) facilitating degeneration, cytoplasmic fragmentation and apoptosis; (4) worsening the disorganization of cortical granules and spindle/chromosomes; and (5) impairing redox potential with increased oxidative stress. In conclusion, FRSOD impairs oocyte resistance to POA, so that stressed oocytes become aged significantly quicker than unstressed controls. Thus, couples wishing to achieve pregnancy should take steps to avoid not only fertilization of aged oocytes but also pregestational stressful life events.


Oocytes , Oogenesis , Female , Mice , Animals , Aging , Ovulation , Oxidative Stress/physiology
8.
Front Cell Dev Biol ; 10: 874374, 2022.
Article En | MEDLINE | ID: mdl-35433692

Most studies on mechanisms by which prenatal stress affects offspring behavior were conducted during late pregnancy using in vivo models; studies on the effect of preimplantation stress are rare. In vivo models do not allow accurate specification of the roles of different hormones and cells within the complicated living organism, and cannot verify whether hormones act directly on embryos or indirectly to alter progeny behavior. Furthermore, the number of anxiety-related miRNAs identified are limited. This study showed that both mouse embryculture with corticosterone (ECC) and maternal preimplantation restraint stress (PIRS) increased anxiety-like behavior (ALB) while decreasing hippocampal expression of glucocorticoid receptor (GR) and brain-derived neurotrophic factor (BDNF) in offspring. ECC/PIRS downregulated GR and BDNF expression by increasing miR-211-5p expression via promoter demethylation of its host gene Trpm1, and this epigenetic cell fate determination was exclusively perpetuated during development into mature hippocampus. Transfection with miR-211-5p mimic/inhibitor in cultured hippocampal cell lines confirmed that miR-211-5p downregulated Gr and Bdnf. Intrahippocampal injection of miR-211-5p agomir/antagomir validated that miR-211-5p dose-dependently increased ALB while decreasing hippocampal GR/BDNF expression. In conclusion, preimplantation exposure to glucocorticoids increased ALB by upregulating miR-211-5p via Trpm1 demethylation, and miR-211-5p may be used as therapeutic targets and biomarkers for anxiety-related diseases.

9.
Biol Reprod ; 106(5): 900-909, 2022 05 17.
Article En | MEDLINE | ID: mdl-35084021

Oocyte aneuploidy is caused mainly by chromosome nondisjunction and/or unbalanced sister chromatid pre-division. Although studies in somatic cells have shown that topoisomerase II (TOP2) plays important roles in chromosome condensation and timely separation of centromeres, little is known about its role during oocyte meiosis. Furthermore, because VP-16, which is a TOP2 inhibitor and induces DNA double strand breaks, is often used for ovarian cancer chemotherapy, its effects on oocytes must be studied for ovarian cancer patients to recover ovarian function following chemotherapy. This study showed that inhibiting TOP2 with either ICRF-193 or VP-16 during meiosis I impaired chromatin condensation, chromosome alignment, TOP2α localization, and caused metaphase I (MI) arrest and first polar body (PB1) abscission failure. Inhibiting or neutralizing either spindle assembly checkpoint (SAC), Aurora B or maturation-promoting factor (MPF) significantly abolished the effect of ICRF-193 or VP-16 on MI arrest. Treatment with ICRF-193 or VP-16 significantly activated MPF and SAC but the effect disappeared when Aurora B was inhibited. Most of the oocytes matured in the presence of ICRF-193 or VP-16 were arrested at MI, and only 11-27% showed PB1 protrusion. Furthermore, most of the PB1 protrusions formed in the presence of ICRF-193 or VP-16 were retracted after further culture for 7 h. In conclusion, TOP2 dysfunction causes MI arrest by activating Aurora B, SAC, and MPF, and it prevents PB1 abscission by promoting chromatin bridges.


Aurora Kinase B , M Phase Cell Cycle Checkpoints , Maturation-Promoting Factor , Animals , Aurora Kinase B/metabolism , Chromatin , DNA Topoisomerases, Type II/genetics , Etoposide , Female , Maturation-Promoting Factor/metabolism , Meiosis , Metaphase , Mice , Oocytes , Polar Bodies , Spindle Apparatus , Topoisomerase II Inhibitors
10.
Sci Rep ; 11(1): 7952, 2021 04 12.
Article En | MEDLINE | ID: mdl-33846458

Pathophysiological mechanisms for depression/anxiety are largely unknown. Evidence for transgenerational transmission of acquired epigenetic marks remains limited. We bred unstressed (US) female mice with adolescently restraint-stressed (RS), social instability-stressed (SI) or US males to produce RS, SI and control F1 offspring, respectively. Compared to controls, while paternal RS decreased anxiety-like behavior (ALB) in both female and male offspring, paternal SI increased ALB only in female offspring. Next-generation sequencing and bioinformatics using RS and SI female offspring identified 5 candidate anxiety-transmitting (CAT) genes; each showed a consistent pattern of DNA methylation from F0 spermatozoa through F1 blastocysts to fetal and adult hippocampi. Further analyses validated 4 CAT genes, demonstrated that paternal SI caused ALB differences between male and female offspring through modifying the CAT genes, and indicated a strong correlation between inflammation and ALB pathogenesis and an important function for intronic DNA methylation in regulating ALB-related genes. In conclusion, this study identified important CAT genes and suggested the possibility that stresses on males might alter offspring's ALB by modifying sperm DNA methylation.


Anxiety/genetics , Behavior, Animal/physiology , High-Throughput Nucleotide Sequencing , Restraint, Physical , Stress, Psychological/genetics , Animals , DNA Methylation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Hippocampus/metabolism , Male , Mice , Phenotype , Protein Interaction Maps/genetics , Reproducibility of Results , Social Behavior , Spermatozoa/metabolism
11.
Reprod Fertil Dev ; 33(3): 229-238, 2021 Feb.
Article En | MEDLINE | ID: mdl-33412088

Although invivo and invitro zearalenone (ZEN) exposure impaired oocyte quality, the mechanisms by which ZEN damages oocytes and the lowest observed effect level remain unclear. Furthermore, although it is known that premature chromatin condensation may occur in oocytes under proapoptotic conditions, whether ZEN exposure compromises oocyte competence by impairing gene transcription by causing premature chromatin condensation remains to be investigated. This study tested the toxic concentrations of invivo ZEN exposure that impair oocyte preimplantation developmental potential (PIDP) and the hypothesis that ZEN exposure compromises oocyte competence by increasing oxidative stress and changing chromatin configuration and the transcription of related genes. We found that invivo treatment of mice (Kunming strain, 8 weeks after birth) with 0.5-1mg kg-1 ZEN daily for 5 days, impaired the PIDP of mouse oocytes, increased oxidative stress, disturbed spindle assembly and chromosome segregation, caused premature chromatin condensation, impaired global gene transcription and disturbed the expression of genes related to oocyte competence, spindle assembly, redox potential and apoptosis. In conclusion, ZEN dose-dependently compromised the competence of mouse oocytes by causing oxidative stress and impairing chromatin configuration and gene transcription.


Blastocyst/drug effects , Chromatin Assembly and Disassembly/drug effects , Gene Expression Regulation, Developmental/drug effects , Oocytes/drug effects , Transcription, Genetic/drug effects , Zearalenone/toxicity , Animals , Apoptosis/drug effects , Blastocyst/metabolism , Blastocyst/pathology , Cells, Cultured , Embryo Culture Techniques , Female , In Vitro Oocyte Maturation Techniques , Mice , Oocytes/metabolism , Oocytes/pathology , Oxidative Stress/drug effects
12.
Biol Reprod ; 103(3): 534-547, 2020 08 21.
Article En | MEDLINE | ID: mdl-32588041

Studies suggested that postovulatory oocyte aging might be prevented by maintaining a high maturation-promoting factor (MPF) activity. Whether AMP-activated protein kinase (AMPK) plays any role in postovulatory oocyte aging is unknown. Furthermore, while activation of AMPK stimulates meiotic resumption in mouse oocytes, it inhibits meiotic resumption in pig and bovine oocytes. Thus, the species difference in AMPK regulation of oocyte MPF activities is worth in-depth studies. This study showed that AMPK activation with metformin or 5-aminoimidazole- 4-carboxamide- 1-beta-d- ribofuranoside and inactivation with compound C significantly increased and decreased, respectively, the activation susceptibility (AS) and other aging parameters in aging mouse oocytes. While AMPK activity increased, MPF activity and cyclic adenosine monophosphate (cAMP) decreased significantly with time post ovulation. In vitro activation and inactivation of AMPK significantly decreased and increased the MPF activity, respectively. MPF upregulation with MG132 or downregulation with roscovitine completely abolished the effects of AMPK activation or inactivation on AS of aging oocytes, respectively. AMPK facilitated oocyte aging with increased reactive oxygen species (ROS) and cytoplasmic calcium. Furthermore, treatment with Ca2+/calmodulin-dependent protein kinase (CaMK) inhibitors significantly decreased AS and AMPK activation. Taken together, the results suggested that AMPK facilitated oocyte aging through inhibiting MPF activities, and postovulatory oocyte aging activated AMPK with decreased cAMP by activating CaMKs via increasing ROS and cytoplasmic calcium.


AMP-Activated Protein Kinases/metabolism , Oocytes/growth & development , Ovulation/physiology , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Culture Media, Conditioned , Cyclic AMP/metabolism , Enzyme Activation , Female , Membrane Potential, Mitochondrial/drug effects , Mesothelin , Metformin/pharmacology , Mice , Pregnancy , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism
13.
J Cell Physiol ; 235(10): 7530-7540, 2020 10.
Article En | MEDLINE | ID: mdl-32100885

Successful use of oocytes from small follicles (SFs) is of great importance for animal embryo production and human in vitro fertilization with reduced hormone-related side effects. How in vitro meiotic arrest maintenance (MAM) increases the competence of oocytes is not clear. In this study, pig oocytes recovered from SF of 1-2 mm and medium-follicles (MF) of 3-6 mm in diameter from abattoir ovaries were treated by various MAM treatments to improve their competence. The results showed that 25 µM roscovitine or 1 mM db-cAMP efficiently blocked germinal vesicle breakdown in both SF and MF oocytes suggesting a similar cyclin-dependent kinase (CDK) 1 level between the two oocyte groups. MAM with 15- and 25-µM roscovitine alone or with 1-mM db-cAMP improved competence of SF and MF oocytes, respectively, with a promoted chromatin configuration transition from surrounded nucleoli (SN) to re-decondensation (RDC) pattern that supported substantial gene transcription. However, MAM with db-cAMP alone or with higher concentrations of roscovitine did not improve oocyte competence, could not support an SN-to-RDC transition, and/or evoked a premature chromatin condensation (PMC) that suppressed gene transcription. Both CDK2 and CDK5 contents were higher (p < .05) in MF than in SF oocytes. It is concluded that the competence of pig oocytes, particularly that of SF oocytes can be improved by MAM using a proper roscovitine concentration that promotes gene transcription by inhibiting CDK5 while letting CDK2 off to prevent PMC.


Cyclin-Dependent Kinases/metabolism , Meiosis/drug effects , Oocytes/drug effects , Oocytes/metabolism , Roscovitine/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin/metabolism , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Female , In Vitro Oocyte Maturation Techniques/methods , Swine , Transcription, Genetic/drug effects
14.
Zygote ; : 1-10, 2020 Jan 14.
Article En | MEDLINE | ID: mdl-31933449

Studies have indicated that psychological stress impairs human fertility and that various stressors can induce apoptosis of testicular cells. However, the mechanisms by which psychological stress on males reduces semen quality and stressors induce apoptosis in testicular cells are largely unclear. Using a psychological (restraint) stress mouse model, we tested whether male psychological stress triggers apoptosis of spermatozoa and spermatogenic cells through activating tumour necrosis factor (TNF)-α signalling. Wild-type or TNF-α-/- male mice were restrained for 48 h before examination for apoptosis and expression of TNF-α and TNF receptor 1 (TNFR1) in spermatozoa, epididymis, seminiferous tubules and spermatogenic cells. The results showed that male restraint significantly decreased fertilization rate and mitochondrial membrane potential, while increasing levels of malondialdehyde, active caspase-3, TNF-α and TNFR1 in spermatozoa. Male restraint also increased apoptosis and expression of TNF-α and TNFR1 in caudae epididymides, seminiferous tubules and spermatogenic cells. Sperm quality was also significantly impaired when spermatozoa were recovered 35 days after male restraint. The restraint-induced damage to spermatozoa, epididymis and seminiferous tubules was significantly ameliorated in TNF-α-/- mice. Furthermore, incubation with soluble TNF-α significantly reduced sperm motility and fertilizing potential. Taken together, the results demonstrated that male psychological stress induces apoptosis in spermatozoa and spermatogenic cells through activating the TNF-α system and that the stress-induced apoptosis in spermatogenic cells can be translated into impaired quality in future spermatozoa.

15.
Aging (Albany NY) ; 10(7): 1745-1757, 2018 Jul 26.
Article En | MEDLINE | ID: mdl-30048240

Although previous studies indicated that cumulus cells (CCs) accelerate oocyte aging by releasing soluble factors, the factors have yet to be characterized. While demonstrating that CCs promoted oocyte aging by releasing soluble Fas ligand (sFasL), our recent study suggested that CCs might secrete other factors to mediate oocyte aging as well. This study tested whether CCs accelerate oocyte aging by secreting tumor necrosis factor (TNF)-α. The results showed that mouse CCs undergoing apoptosis released soluble TNF-α (sTNF-α) during in vitro aging. While ethanol activation rates were higher, the maturation-promoting factor (MPF) activity was lower significantly after culture of cumulus-denuded oocytes (DOs) in medium conditioned with CCs for 36 h than in medium conditioned for 24 h. Aging mouse oocytes expressed TNF-receptor 1. The CCs released equal amounts of sTNF-α and sFasL during aging in vitro, and the TNF-α-knockdown CCs secreted less sFasL than the control CCs did. Treatment of DOs in vitro with sTNF-α significantly accelerated their aging. The aging-promoting effect of sTNF-α was significantly reduced in TNF-α-knocked-down CCs and in CCs from the TNF-α-knockout mice. It is concluded that mouse CCs accelerate oocyte aging by secreting sTNF-α as well as sFasL.


Cellular Senescence/physiology , Cumulus Cells/metabolism , Oocytes/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Chromosomes , Coculture Techniques , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Mesothelin , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovulation/physiology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Spindle Apparatus/physiology
16.
J Cell Physiol ; 233(9): 6952-6964, 2018 09.
Article En | MEDLINE | ID: mdl-29336483

In previous studies on glucose metabolism during in vitro maturation, intact cumulus-oocyte complexes (COCs) were treated with enzyme inhibitors/activators. Because inhibitors/activators may have non-specificity and/or toxicity, and culture of COCs cannot differentiate whether glucose metabolism of cumulus cells (CCs) or that of the oocyte supports oocyte maturation, results from the previous studies must be verified by silencing genes in either CCs or cumulus-denuded oocytes (DOs). In this study, RNAi was adopted to specify the effects of glucose metabolism in CCs or DOs on oocyte maturation. Although silencing either glyceraldehyde 3-phosphate dehydrogenase (GAPDH) or glucose-6-phosphate dehydrogenase (G6PD) genes in CCs significantly decreased competence of the cocultured DOs, silencing G6PD impaired competence to a greater extent. While silencing G6PD or GAPDH of CCs decreased glutathione and ATP contents of cocultured DOs to similar extents, silencing G6PD increased oxidative stress as well. Analysis on metabolite contents and oxidative stress index and culture of DOs in medium conditioned with gene-silenced CCs indicated that CCs supported oocyte maturation by releasing glucose metabolites. Silencing mitochondrial pyruvate carrier 1 or NADH dehydrogenase (ubiquintone) flavoprotein 1 of DOs significantly impaired their maturation. The results have unequivocally confirmed that CCs promote oocyte maturation by releasing glucose metabolites from both pentose phosphate pathway (PPP) and glycolysis. Pyruvate is transferred into DOs by mitochondrial pyruvate carrier (MPC) and utilized through mitochondrial electron transport to support maturation.


Glucose/metabolism , In Vitro Oocyte Maturation Techniques , Oocytes/cytology , Oocytes/metabolism , RNA Interference , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Down-Regulation/drug effects , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Glucosephosphate Dehydrogenase/metabolism , Glutathione/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glycolysis/drug effects , Mice , NADP/metabolism , Oocytes/drug effects , Oxidation-Reduction , Pentose Phosphate Pathway/drug effects , Proprotein Convertase 1/metabolism , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
17.
Biol Reprod ; 98(2): 218-226, 2018 02 01.
Article En | MEDLINE | ID: mdl-29267849

Mechanisms for postovulatory aging (POA) of oocytes and for spontaneous activation (SA) of rat oocytes are largely unknown. Expression of calcium-sensing receptor (CaSR) in rat oocytes and its role in POA remain unexplored. In this study, expression of CaSR in rat oocytes aging for different times was detected by immunofluorescence microscopy, and western blotting and the role of CaSR in POA was determined by observing the effects of regulating its activity on SA susceptibility and cytoplasmic calcium levels. The results showed that CaSR was expressed in rat oocytes. Oocytes recovered 19 h post human chorionic gonadotropin (hCG) injection were more susceptible to SA and expressed more functional CaSR than oocytes recovered 13 h after hCG injection, although both expressed the same level of total CaSR protein. Treatment with CaSR antagonist significantly suppressed cytoplasmic calcium elevation and SA of oocytes. Activation of Na-Ca2+ exchanger with NaCl inhibited SA to a greater extent than suppression of CaSR with NPS-2143, suggesting that calcium sources other than CaSR-controlled channels contributed to the elevation of cytoplasmic calcium. Treatment with T- or L-type calcium channel blockers significantly reduced SA. Suppression of all calcium channels tested reduced SA to minimum. It is concluded that the level of CaSR functional dimer protein, but not that of the total CaSR protein, was positively correlated with the SA susceptibility during POA of rat oocytes confirming that CaSR is involved in POA regulation. Blocking multiple calcium channels might be a better choice for efficient control of SA in rat oocytes.


Oocytes/metabolism , Ovulation/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Chorionic Gonadotropin/pharmacology , Female , Naphthalenes/pharmacology , Oocytes/drug effects , Ovulation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism
18.
Aging (Albany NY) ; 9(4): 1186-1201, 2017 04.
Article En | MEDLINE | ID: mdl-28394765

In this study, microRNA (miRNA) profiles in postovulatory aging mouse oocytes were analyzed by microarray screening and RT-qPCR. Hierarchical cluster analysis on the microarray data and KEGG pathway enrichment analysis on the mRNAs targeted by differentially expressed (DE) miRNAs between two adjacent egg-ages suggest that while only a mild alteration in miRNA expression occurred from 13 to 18 h, a great change took place from 18 to 24 h post hCG injection. Theoretical exploration on functions of the predicted target genes suggest that KEGG pathways enriched by 13-18 h DE miRNAs are correlated with early events of oocyte aging while pathways most enriched by 18-24 h or 24-30 h DE miRNAs are correlated with the late symptoms of aged oocytes. Experimental verification on functions of the key proteins predicted by the KEGG analysis and injection of miR-98 mimics or inhibitors further confirmed that miRNAs played stimulatory/inhibitory roles in postovulatory oocyte aging. In conclusion, marked changes in miRNA expression are associated with significant alterations in function and morphology of postovulatory aging oocytes.


Gene Expression Regulation, Developmental/genetics , MicroRNAs/biosynthesis , Oocytes/physiology , Animals , Calcium/metabolism , Caspase 3/biosynthesis , Caspase 3/genetics , Computational Biology , Female , Gene Expression Profiling , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Microarray Analysis , Oocytes/metabolism , Ovulation , Polymerase Chain Reaction , Signal Transduction/genetics
19.
Biol Reprod ; 96(5): 1019-1030, 2017 May 01.
Article En | MEDLINE | ID: mdl-28419184

Although in vitro exposure to physiological concentrations of glucorticoids did not affect maturation of mouse oocytes, it significantly inhibited nuclear maturation of pig oocytes. Studies on this species difference in oocyte sensitivity to glucocorticoids will contribute to our understanding of how stress/glucocorticoids affect oocytes. We showed that glucorticoid receptors (NR3C1) were expressed in both oocytes and cumulus cells (CCs) of both pigs and mice; however, while cortisol inhibition of oocyte maturation was overcome by NR3C1 inhibitor RU486 in pigs, it could not be relieved by RU486 in mice. The mRNA level of 11ß-hydroxysteroid dehydrogenase 1 (HSD11B1) was significantly higher than that of HSD11B2 in pig cumulus-oocyte complexes (COCs), whereas HSD11B2 was exclusively expressed in mouse COCs. Pig and mouse cumulus-denuded oocytes (DOs) expressed HSD11B2 predominantly and exclusively, respectively. In the presence of cortisol, although inhibiting HSD11B2 decreased maturation rates of COCs in both species, inhibiting HSD11B1 improved maturation of pig COCs while having no effect on mouse COCs. Cortisol-cortisone interconversion observation confirmed high HSD11B1 activities in pig oocytes but none in mouse oocytes, a higher HSD11B2 activity in mouse than in pig oocytes, and a rapid cortisol-cortisone interconversion in pig COCs catalyzed by HSD11B1 from CCs and HSD11B2 from DOs. In conclusion, the species difference in glucocorticoid sensitivity between pig and mouse oocytes is caused by their different contents/ratios of HSD11B1 and HSD11B2, which maintain different concentrations of active glucocorticoids. While cortisol inhibited pig oocytes by interacting with NR3C1, glucocorticoid suppression of mouse oocytes was apparently not mediated by NR3C1.


Glucocorticoids/pharmacology , Oocytes/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Animals , Cortisone/metabolism , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Hydrocortisone/metabolism , Mice , Mifepristone/pharmacology , Oogenesis , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Species Specificity , Sus scrofa
20.
Reprod Sci ; 23(9): 1148-57, 2016 09.
Article En | MEDLINE | ID: mdl-26880768

AIM: Although previous studies found that 1-time acute stress applied during follicle maturation impaired oocyte competence, it is unknown whether repeated chronic stress, which is known to cause animal behavioral adaptation, would damage oocytes when applied during follicle growth. METHODS AND RESULTS: In this study, female mice were exposed to repeated restraint stress (RRS) or unpredictable stress (UPS) for different days before equine chorionic gonadotropin injection to initiate oocyte prematuration development and to observe effects of different stressors on oocytes in the growing follicles. The results showed that although oocyte pre- and postimplantation development was unaffected when mice were exposed to RRS or UPS once a day for 4 days, development was impaired when mice were exposed to RRS for 8 or more days or to UPS twice a day for 4 days (4 × 2). The 4 × 2 UPS caused more oxidative stress in oocytes and severer apoptosis in antral follicles than did the 4-day RRS. The RRS mice were stressed consistently from days 1 to 23 of restraint, and the stress that a mouse had 4 × 2 UPS was severer than that from 4-day RRS. CONCLUSION: The results suggest that (1) the degree that a stress damages oocytes is the product of duration × severity of the stress; (2) RRS impaired oocyte developmental potential through cumulative effects on growing follicles; and (3) preantral follicles were not as sensitive to stress as antral follicles were.


Oocytes/physiology , Ovarian Follicle/growth & development , Ovarian Follicle/physiopathology , Stress, Psychological/physiopathology , Animals , Anxiety/etiology , Anxiety/physiopathology , Apoptosis , Body Weight , Female , Glucocorticoids/blood , Glutathione/metabolism , Hydrocortisone/blood , Mice , Oocytes/metabolism , Restraint, Physical , Stress, Psychological/complications
...