Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 236(10): 6948-6962, 2021 10.
Article in English | MEDLINE | ID: mdl-33682931

ABSTRACT

ACSS1/2 converts acetate into acetyl-coenzyme A, which contributes to histone acetylation in the mitochondria and cytoplasm. Zygotic genome activation (ZGA) is critical for embryo development involving drastic histone modification. An efficient crRNAs-Cas13a targeting strategy was employed to investigate the ACSS1/2 function during ZGA. The results showed that nuclear accumulation of ACSS1 and ACSS2 occurs during ZGA. Knockdown of ACSS1/2 did not affect blastocyst formation when using a normal medium. On culturing embryos in a medium with acetate and no pyruvate (-P + Ace), knockdown of ACSS1 did not affect histone acetylation levels but significantly reduced ATP levels, whereas knockdown of ACSS2 significantly reduced histone acetylation levels in porcine embryos. Inhibition of fatty acid beta-oxidation by etomoxir significantly reduced ATP levels, which could be restored by acetate. The histone acetylation levels in the ACSS1 and ACSS2 knockdown groups both decreased considerably after etomoxir treatment. Moreover, acetate showed dose-dependent effects on SIRT1 and SIRT3 levels when under metabolic stress. The C-terminus of ACSS1 regulated the nuclear translocation. In conclusion, ACSS1/2 helps to maintain ATP and histone acetylation levels in porcine early embryos under metabolic stress during ZGA.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetates/metabolism , Acetyl Coenzyme A/metabolism , Energy Metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Protein Processing, Post-Translational , Zygote/enzymology , Acetate-CoA Ligase/genetics , Acetylation , Adenosine Triphosphate/metabolism , Animals , Embryo Culture Techniques , Parthenogenesis , Sirtuin 1/metabolism , Sirtuin 3/metabolism , Sus scrofa
2.
Microsc Microanal ; 27(2): 385-391, 2021 04.
Article in English | MEDLINE | ID: mdl-33413706

ABSTRACT

The cytoskeleton plays an orchestrating role in polarized cell growth. Microtubules (MTs) not only play critical roles in chromosome alignment and segregation but also control cell shape, division, and motility. A member of the plus-end tracking proteins, end-binding protein 1 (EB1), regulates MT dynamics and plays vital roles in maintaining spindle symmetry and chromosome alignment during mitosis. However, the role of EB1 in mouse oocyte meiosis remains unknown. Here, we examined the localization patterns and expression levels of EB1 at different stages. EB1 protein level was found to be stable during meiosis. EB1 mainly localized along the spindle and had a similar localization pattern as that of α-tubulin. The EB1 protein was degraded with a Trim-Away method, and the results were further confirmed with western blotting and immunofluorescence. At 12 h of culture after EB1 knockdown (KD), a reduced number of mature MII oocytes were observed. EB1 KD led to spindle disorganization, chromosome misalignment, and missegregation; ß-catenin protein binds to actin via the adherens junctional complex, which was significantly reduced in the EB1 KD oocytes. Collectively, we propose that the impairment of EB1 function manipulates spindle formation, thereby promoting chromosomal loss, which is expected to fuel aneuploidy and possibly fertilization failure.


Subject(s)
Meiosis , Spindle Apparatus , Animals , Chromosomes , Mice , Microtubules , Oocytes
3.
J Cell Sci ; 133(23)2020 12 07.
Article in English | MEDLINE | ID: mdl-33148610

ABSTRACT

Activator of G-protein signaling 3 (AGS3, also known as GPSM1) regulates the trans-Golgi network. The AGS3 GoLoco motif binds to Gαi and thereby regulates the transport of proteins to the plasma membrane. Compaction of early embryos is based on the accumulation of E-cadherin (Cdh1) at cell-contacted membranes. However, how AGS3 regulates the transport of Cdh1 to the plasma membrane remains undetermined. To investigate this, AGS3 was knocked out using the Cas9-sgRNA system. Both trans-Golgi network protein 46 (TGN46, also known as TGOLN2) and transmembrane p24-trafficking protein 7 (TMED7) were tracked in early mouse embryos by tagging these proteins with a fluorescent protein label. We observed that the majority of the AGS3-edited embryos were developmentally arrested and were fragmented after the four-cell stage, exhibiting decreased accumulation of Cdh1 at the membrane. The trans-Golgi network and TMED7-positive vesicles were also dispersed and were not polarized near the membrane. Additionally, increased Gαi1 (encoded by GNAI1) expression could rescue AGS3-overexpressed embryos. In conclusion, AGS3 reinforces the dynamics of the trans-Golgi network and the transport of TMED7-positive cargo containing Cdh1 to the cell-contact surface during early mouse embryo development.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/genetics , Protein Transport , trans-Golgi Network , Animals , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Mice , Signal Transduction , trans-Golgi Network/metabolism
4.
Microsc Microanal ; 26(2): 287-296, 2020 04.
Article in English | MEDLINE | ID: mdl-32036801

ABSTRACT

Connexin 43 (CX43) is a component of gap junctions. The lack of functional CX43 induces oxidative stress, autophagy, and apoptosis in somatic cells. However, the role of CX43 in the early development of porcine embryos is still unknown. Thus, the aim of this study was to investigate the role of CX43, and its underlying molecular mechanisms, on the developmental competence of early porcine embryos. We performed CX43 knockdown by microinjecting dsRNA into parthenogenetically activated porcine parthenotes. The blastocyst development rate and the total number of cells in the blastocysts were significantly reduced by CX43 knockdown. Results from FITC-dextran assays showed that CX43 knockdown significantly increased membrane permeability. ZO-1 protein was obliterated in CX43 knockdown blastocysts. Mitochondrial membrane potential and ATP production were significantly reduced following CX43 knockdown. Reactive oxygen species (ROS) levels were significantly increased in the CX43 knockdown group compared to those in control embryos. Moreover, CX43 knockdown induced autophagy and apoptosis. Our findings indicate that CX43 is essential for the development and preimplantation of porcine embryos and maintains mitochondrial function, cell junction structure, and cell homeostasis by regulating membrane permeability, ROS generation, autophagy, and apoptosis in early embryos.


Subject(s)
Connexin 43/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Mitochondria/metabolism , Animals , Apoptosis , Autophagy , Blastocyst/metabolism , Gene Knockdown Techniques , Intercellular Junctions , Membrane Potential, Mitochondrial/physiology , Oocytes , Oxidative Stress , Reactive Oxygen Species , Swine
5.
Aging (Albany NY) ; 12(2): 1256-1271, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31958774

ABSTRACT

Ubiquinol-10, the reduced form of coenzyme Q10, protects mammalian cells from oxidative damage and enhances mitochondrial activity. However, the protective effect of ubiquinol-10 on mammalian oocytes is not well understood. In this study, we investigated the effect of ubiquinol-10 on porcine oocytes during postovulatory aging. Metaphase II oocytes were selected as fresh oocytes and further cultured for 48 h with different concentrations of ubiquinol-10 (0-400 µM) in vitro as a postovulatory aging model. After choosing the optimal concentration of ubiquinol-10 (100 µM) that maintained oocyte morphology and developmental competence during the progression of aging, the oocytes were randomly divided into five groups: fresh, control-24 h, ubiquinol-24 h, control-48 h, and ubiquinol-48 h. The results revealed that ubiquinol-10 significantly prevented aging-induced oxidative stress, GSH reduction, cytoskeleton impairment, apoptosis, and autophagy. Mitochondrial biogenesis (SIRT1 and PGC-1α) and mitophagy (PINK1 and PARKIN)-related proteins were decreased during aging. Addition of ubiquinol-10 prevented the aging-induced reduction of these proteins. Consequently, although mitochondrial content was decreased, the number of active mitochondria and ATP level were significantly increased upon treatment with ubiquinol-10. Thus, ubiquinol-10 has beneficial effects on porcine postovulatory aging oocytes owing to its antioxidant properties and ability to promote mitochondrial renewal.


Subject(s)
Cellular Senescence/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Oocytes/drug effects , Oocytes/physiology , Ubiquinone/analogs & derivatives , Aging , Animals , Apoptosis , Embryonic Development , Mitophagy , Ovulation , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Swine , Ubiquinone/pharmacology
6.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118648, 2020 04.
Article in English | MEDLINE | ID: mdl-31935425

ABSTRACT

Porcine zygotic genome activation (ZGA) occurs along with global epigenetic remodeling at the 4-cell stage. These processes are regulated by histone acetylation, which requires acetyl-coenzyme A (CoA). Pyruvate dehydrogenase complex (PDC) is a crucial enzyme in glucose metabolism that converts pyruvate into acetyl-CoA. In mammalian cells, acetyl-CoA is produced by pyruvate dehydrogenase alpha 1 (PDHA1) translocated into the nucleus in special conditions. To determine whether zygotic PDHA1 plays a critical role in promoting histone acetylation during ZGA, a CRISPR/Cas9 genome editing system using multiple guide RNAs was employed to generate a PDHA1-targeted parthenogenetic embryo model. Results of immunofluorescent staining showed that the nuclear accumulation of PDHA1 during ZGA was significantly inhibited by PDHA1 targeting. Meanwhile, the 4-cell arrest rate significantly increased at 72 h after activation, indicating impeded embryonic development. In addition, nuclear histone acetylation significantly decreased when PDHA1 was targeted, and quantitative PCR showed that expression of several zygotic genes was significantly decreased in the PDHA1-targeting group compared to the control group. Overexpression of PDHA1 recovered the nuclear PDHA1, H3K9Ac and H3K27Ac and EIF1A expression levels. Moreover, the 5-to-8-cell-stage embryo development rate was only partially rescued. In conclusion, expression of zygotic origin PDHA1 contributes to porcine ZGA by maintaining histone acetylation in porcine embryos.


Subject(s)
Cell Nucleus/enzymology , Embryonic Development/genetics , Histones/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Zygote/enzymology , Acetylation , Animals , CRISPR-Cas Systems , Embryo, Mammalian/enzymology , Embryo, Mammalian/metabolism , Gene Editing , Gene Expression , Genome , Pyruvate Dehydrogenase (Lipoamide)/genetics , Swine , Zygote/metabolism
7.
J Pineal Res ; 68(2): e12627, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31773776

ABSTRACT

Melatonin, a major hormone of the pineal gland, exerts many beneficial effects on mitochondria. Several studies have shown that melatonin can protect against toxin-induced oocyte quality impairment during maturation. However, there is little information regarding the beneficial effects of melatonin on toxin-exposed early embryos, and the mechanisms underlying such effects have not been determined. Rotenone, a chemical widely used in agriculture, induces mitochondrial toxicity, therefore, damaging the reproductive system, impairing oocyte maturation, ovulation, and fertilization. We investigated whether melatonin attenuated rotenone exposure-induced impairment of embryo development by its mitochondrial protection effect. Activated oocytes were randomly assigned to four groups: the control, melatonin treatment, rotenone-exposed, and "rotenone + melatonin" groups. Treatment with melatonin abrogated rotenone-induced impairment of embryo development, mitochondrial dysfunction, and ATP deficiency, and significantly decreased oxidative stress and apoptosis. Melatonin also increased SIRT1 and PGC-1α expression, which promoted mitochondrial biogenesis. SIRT1 knockdown or pharmacological inhibition abolished melatonin's ability to revert rotenone-induced impairment. Thus, melatonin rescued rotenone-induced impairment of embryo development by reducing ROS production and promoting mitochondrial biogenesis. This study shows that melatonin rescues toxin-induced impairment of early porcine embryo development by promoting mitochondrial biogenesis.


Subject(s)
Embryo, Mammalian , Embryonic Development/drug effects , Melatonin/pharmacology , Mitochondria , Mitochondrial Diseases , Rotenone/adverse effects , Animals , Embryo, Mammalian/embryology , Embryo, Mammalian/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/embryology , Mitochondrial Diseases/prevention & control , Rotenone/pharmacology , Swine
8.
Toxicol In Vitro ; 61: 104635, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31479719

ABSTRACT

Thiamethoxam (TMX) is a neonicotinoid insecticide, the residues of which have been detected on various crops. In addition to its specific acetylcholine toxicity to insects, TMX was also found to be toxic to mammals. Moreover, oocytes are vulnerable to reactive oxygen species (ROS). Excessive ROS production can override antioxidant defenses and produce oxidative stress and DNA damage that trigger apoptosis and necrosis in organisms. In this study, we exposed bovine oocytes to TMX during maturation. Microscopic examination showed that 1.6 mM TMX significantly inhibited maturation at the germinal vesicle (GV) and metaphase I (MI) stages. Immunofluorescence staining and enzyme activity analysis revealed that TMX induced a reduction in CDC25 and CDC2 activity. Furthermore, time-lapse tracking and immunofluorescence staining indicated the maintenance of cyclin B in the cytoplasm, persistence of Bub3 at kinetochores, and absence of actin caps after TMX-exposed oocytes reached the MI stage. In addition, metaphase II (MII) oocytes exposed to TMX showed disordered chromosomes and spindles. These oocytes accumulated excess ROS and showed significantly decreased mitochondrial membrane potential and increased apoptotic signals. Parthenogenetic embryos from these oocytes showed decreased percentages of morulae and blastocysts. These results indicate that TMX delays bovine oocyte progression to MI stage, blocks them at the MI stage, triggers disordered chromosomes and spindles at MII stage, and ultimately results in MII oocytes with poor cleavage ability and inhibited development to morulae and blastocysts.


Subject(s)
Insecticides/toxicity , Oocytes/drug effects , Thiamethoxam/toxicity , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cattle , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Female , Meiosis/drug effects , Oocytes/cytology , Oocytes/metabolism , Reactive Oxygen Species/metabolism , cdc25 Phosphatases/metabolism
9.
Apoptosis ; 24(9-10): 718-729, 2019 10.
Article in English | MEDLINE | ID: mdl-31240517

ABSTRACT

Fipronil (FPN) is a widely used phenylpyrazole pesticide that can kill pests by blocking γ-aminobutyric acid (GABA)-gated chloride channels. In addition, there are lack of studies on the effects of FPN on the female mammalian gametes. In this study, porcine oocytes were used to investigate the effects of FPN on the oocyte maturation process. The results showed that the first polar body extrusion rate significantly decreased (100 µM FPN vs. control, 18.64 ± 2.95% vs. 74.90 ± 1.50%, respectively), and oocytes were arrested at the germinal vesicle stage in 100 µM FPN group. Meanwhile, the FPN caused a significant increase in reactive oxygen species (ROS) levels and severe DNA damage inside the oocytes. Furthermore, apoptosis was enhanced along with decreases in mitochondrial membrane potential, BCL-xL, and the release of cytochrome C in FPN-treated group. Additionally, low CDK1 activity and delayed cyclin B1 degradation during germinal vesicle breakdown were found in the FPN-treated group, which resulted from the activation of ATM-P53-P21 pathway. In conclusion, FPN induces apoptosis and cell cycle arrest in porcine oocyte maturation because of increased ROS levels and DNA damage. This suggests that the FPN in the environment may have potential detrimental effects on the female mammalian reproductive system.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Oocytes/drug effects , Pyrazoles/pharmacology , Animals , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Cyclin B1/drug effects , Cytochromes c/drug effects , Cytochromes c/metabolism , DNA Damage/drug effects , Female , In Vitro Techniques , Oocytes/cytology , Oogenesis/drug effects , Pesticides/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Swine , bcl-X Protein/drug effects , bcl-X Protein/metabolism
10.
FASEB J ; 33(7): 7882-7895, 2019 07.
Article in English | MEDLINE | ID: mdl-30897005

ABSTRACT

Phosphatase and tensin homolog-induced kinase 1 (PINK1) on the outer membranes of impaired mitochondria promotes mitophagy and regulates mitochondrial morphology. Mammalian oocytes and early embryos are mitochondria rich, but mitochondrial dynamics during preimplantation embryo development is not well-studied. To investigate whether PINK1 is required for mitochondrial dynamics in porcine preimplantation embryos, gene knockdown and inhibitors were used, and mitochondrial dynamics were observed by transmission electron microscopy. PINK1 knockdown significantly impaired blastocyst formation and quality, induced mitochondrial elongation and swelling, and reduced mitochondrial DNA copy number. PINK1 knockdown-induced mitochondrial elongation caused mitochondrial dysfunction, oxidative stress, and ATP deficiency, significantly increasing autophagy and apoptosis. Profission dynamin-related protein 1 overexpression prevented PINK1 knockdown-induced impairment of embryo development, mitochondrial elongation, and dysfunction. Thus, PINK1 promotes mitochondrial fission in porcine preimplantation embryos.-Niu, Y.-J., Nie, Z.-W., Shin, K.-T., Zhou, W., Cui, X.-S. PINK1 regulates mitochondrial morphology via promoting mitochondrial fission in porcine preimplantation embryos.


Subject(s)
Blastocyst/physiology , Mitochondria/ultrastructure , Mitochondrial Dynamics/physiology , Protein Kinases/physiology , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Autophagy , Blastocyst/metabolism , Dynamins/genetics , Dynamins/physiology , Embryonic Development , Gene Dosage , Gene Knockdown Techniques , Genes, Mitochondrial , In Vitro Oocyte Maturation Techniques , Membrane Potential, Mitochondrial , Microinjections , Parthenogenesis , Protein Kinases/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Recombinant Proteins , Sus scrofa
11.
J Cell Physiol ; 234(6): 8963-8974, 2019 06.
Article in English | MEDLINE | ID: mdl-30317618

ABSTRACT

Spindlin 1 (SPIN1), which contains Tudor-like domains, regulates maternal transcripts via interaction with a messenger RNA (mRNA)-binding protein. SPIN1 is involved in tumorigenesis in somatic cells and is highly expressed in cancer cells. Nevertheless, the role of SPIN1 in porcine oocyte maturation remains totally unknown. To explore the function of SPIN1 in porcine oocyte maturation, knockdown, and overexpression techniques were used. SPIN1 mRNA was identified in maternal stages ranging from GV to MII. SPIN1 was localized in the cytoplasm and to chromosomes during meiosis. SPIN1 knockdown accelerated first polar body extrusion. Oocytes with overexpressed SPIN1 were arrested at the MI stage. SPIN1 depletion caused meiotic spindle defects and chromosome instability. The BUB3 signal was investigated, confirming that SPIN1 affects the stability of Bub3 mRNA as well as BUB3 expression. Further, overexpression of SPIN1 inhibited the degradation and regulation of G2/mitotic-specific cyclin-B1. In summation, SPIN1 regulates the meiotic cell cycle by modulating the activation of the spindle assembly checkpoint.


Subject(s)
Anaphase , Cell Cycle Proteins/metabolism , Metaphase , Microtubule-Associated Proteins/metabolism , Oocytes/metabolism , Phosphoproteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Spindle Apparatus/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosome Segregation , Female , Gene Expression Regulation, Developmental , In Vitro Oocyte Maturation Techniques , Microtubule-Associated Proteins/genetics , Phosphoproteins/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Signal Transduction , Sus scrofa , Time Factors
12.
Cell Signal ; 53: 294-303, 2019 01.
Article in English | MEDLINE | ID: mdl-30145217

ABSTRACT

Thiamethoxam (TMX) is a neonicotinoid insecticide. It has specific high toxicity to insects. Residues of TMX have been detected in various crops. Early embryo quality is vital for fertility. Excessive production of reactive oxygen species (ROS) can override embryonic antioxidant defenses, producing oxidative stress that triggers apoptosis, necrosis, and/or permanent DNA damage responses in the early embryo. Comparative studies have indicated that TMX hepatotoxicity is significant in mammals in acute tests, but little is known about accumulated chronic toxicity in early embryonic development. Porcine embryos were obtained here by the parthenogenetic activation of meiosis II oocytes and cultured in the PZM-5 medium with or without TMX. These embryos were evaluated by various methods. The expansion and hatching of blastocysts treated with TMX decreased by 21.73% and 16.71%, respectively, as compared with controls. In an analysis of 5-bromo-2-deoxyuridine (BrdU) incorporation, the rate of cell proliferation was 44.33% lower as compared with expanded blastocysts of the control group. ROS and γH2AX levels were higher in the TMX group than in the control group. Real-time reverse-transcription polymerase chain reaction showed that Sod1 expression increased and the expression of Mnsod, Gpx1, Igta5, and Cox2 decreased. A CDK1 kinase assay revealed that maturation-promoting factor (MPF) activity diminished by 31.41% in expanding blastocysts. In conclusion, these results suggest that TMX inhibits blastocyst expansion and hatching by ROS-induced DNA damage checkpoint activation, which inhibits the activation of MPF and cell cycle progression in porcine blastocysts.


Subject(s)
Blastocyst/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Insecticides/adverse effects , Reactive Oxygen Species/metabolism , Thiamethoxam/adverse effects , Animals , Blastocyst/cytology , Blastocyst/metabolism , Embryonic Development/drug effects , Female , Pregnancy , Swine/embryology
13.
Cell Cycle ; 17(21-22): 2436-2446, 2018.
Article in English | MEDLINE | ID: mdl-30382773

ABSTRACT

SURVIVIN is an essential chromosomal passenger complex (CPC) subunit and participates in cell division. In this study, we used porcine oocyte as a model to investigate the roles of Survivin during porcine oocyte maturation. Survivin was highly expressed in germinal vesicle (GV) and germinal vesicle breakdown (GVBD) stages oocytes, mainly localized in the GV at GV stage and on the chromosomes after GVBD. We have used RNA interference to specifically deplete Survivin in oocytes during in vitro maturation (IVM). Immunofluorescence assay showed that Survivin-depleted oocytes failed to produce polar body in meiosisⅠ (failed to complete cytokinesis), and they were arrested in metaphaseⅠwith misaligned chromosomes. The homologous chromosomes in Survivin-depleted oocytes could not be separated normally. Moreover, both the phosphorylation levels of Aurora B and the mRNA level of Mad2L1 related to spindle assembly checkpoint (SAC) was decreased in Survivin-depleted oocytes, which thus inhibited the degradation of Cyclin B1 (CCNB1) to complete meiosis. Taken together, we conclude that Survivin is an important mediator of centromere and midbody docking of Aurora-B as well as its activity and regulates SAC and MPF activity during meiosis in porcine oocytes.


Subject(s)
Aurora Kinase B/metabolism , Chromosome Segregation , Meiosis , Oocytes/enzymology , Survivin/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Phosphorylation , Signal Transduction , Spindle Apparatus/enzymology , Spindle Apparatus/genetics , Survivin/genetics , Sus scrofa
14.
Microsc Microanal ; 24(1): 29-37, 2018 02.
Article in English | MEDLINE | ID: mdl-29485024

ABSTRACT

MicroRNA (miR)-29b plays a crucial role during somatic cell reprogramming. The aim of the current study was to explore the effects of miR-29b on the developmental competence of bovine somatic cell nuclear transfer (SCNT) embryos, as well as the underlying mechanisms of action. The expression level of miR-29b was lower in bovine SCNT embryos at the pronuclear, 8-cell, and blastocyst stages compared with in vitro fertilized embryos. In addition, miR-29b regulates the expression of DNA methyltransferases (Dnmt3a/3b and Dnmt1) in bovine SCNT embryos. We further investigated SCNT embryo developmental competence and found that miR-29b overexpression during bovine SCNT embryonic development does not improve developmental potency and downregulation inhibits developmental potency. Nevertheless, the quality of bovine SCNT embryos at the blastocyst stage improved significantly. The expression of pluripotency factors and cellular proliferation were significantly higher in blastocysts from the miR-29b overexpression group than the control and downregulation groups. In addition, outgrowth potential in blastocysts after miR-29b overexpression was also significantly greater in the miR-29b overexpression group than in the control and downregulation groups. Taken together, these results demonstrated that miR-29b plays an important role in bovine SCNT embryo development.


Subject(s)
Blastocyst/metabolism , Cell Nucleus/metabolism , MicroRNAs/metabolism , Animals , Cattle , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Fluorescent Antibody Technique , Nuclear Transfer Techniques , Oocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Environ Mol Mutagen ; 59(3): 223-233, 2018 04.
Article in English | MEDLINE | ID: mdl-29285797

ABSTRACT

Sodium fluoride (NaF) is used as a medicine to prevent tooth decay; however, excessive NaF could cause a pathological damage to the health. Recent studies showed that NaF impaired mouse oocyte maturation, included of abnormal spindle configuration, actin cap formation, cortical granule-free domain formation, and the following development after fertilization. However, few studies used large animals as models to study the toxicology of NaF on oocytes maturation. We proposed a hypothesis that NaF would affect the nuclear and cytoplasmic maturation of porcine oocytes and DNA methylation pattern of imprinted genes in oocytes. Our results showed that NaF affected cumulus expansion, polar body emission, spindle morphology, cortical granule distribution, early apoptosis, and the following development after parthenogenetic activation during porcine oocyte maturation. Moreover, NaF increased the DNA methylation of NNAT and decreased its expression, which disturbed the glucose transport in oocytes. These results suggest that NaF impairs the porcine oocytes maturation epigenetically, which provides a new toxicological mechanism of NaF on the oocyte maturation. Environ. Mol. Mutagen. 59:223-233, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
DNA Methylation , Gene Expression Regulation/drug effects , Glucose/metabolism , In Vitro Oocyte Maturation Techniques/methods , Nerve Tissue Proteins/genetics , Oocytes/metabolism , Sodium Fluoride/pharmacology , Animals , Cariostatic Agents/pharmacology , Cells, Cultured , Female , Nerve Tissue Proteins/metabolism , Oocytes/cytology , Oocytes/drug effects , Oogenesis/drug effects , Swine
16.
Sci Rep ; 7(1): 17082, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213094

ABSTRACT

Excessive long-term fluoride intake is associated with several health problems, including infertility. However, limited information is available on the toxic effects of fluoride exposure on the female reproductive system, especially oocyte maturation. In this study, we investigated the toxic effect of sodium fluoride (NaF) exposure on porcine oocyte maturation and its possible underlying mechanisms. Our results showed that NaF exposure during porcine oocyte maturation inhibited cumulus cell expansion and impaired polar body extrusion. Cell cycle analysis showed that NaF exposure blocked meiotic resumption, disturbed spindle dynamics, disrupted chromosome separation, and increased aneuploidy in porcine oocytes. Moreover, NaF exposure disturbed mitochondrial function, triggered DNA damage response, and induced early apoptosis in porcine oocytes. NaF exposure also induced oxidative stress, decreased GSH level, and increased cathepsin B activity in and impaired the further development potential of porcine oocytes, as indicated by a decrease in blastocyst formation rate, increase in apoptosis, and inhibition of cell proliferation. Together, these results indicate that NaF exposure impairs the maturation capacity of porcine oocytes by inhibiting cumulus cell expansion, disturbing cytoskeletal dynamics, and blocking nuclear and cytoplasmic maturation, thus decreasing the quality and affecting the subsequent embryonic development potential of porcine oocytes.


Subject(s)
Oogenesis/drug effects , Sodium Fluoride/pharmacology , Aneuploidy , Animals , Apoptosis/drug effects , Cathepsin B/metabolism , Chromosome Segregation/drug effects , Cumulus Cells/cytology , Cumulus Cells/metabolism , DNA Damage/drug effects , Female , Glutathione/metabolism , Histones/metabolism , Meiosis/drug effects , Oocytes/cytology , Oocytes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Swine
17.
Sci Rep ; 7(1): 16992, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29208995

ABSTRACT

C-Phycocyanin (CP) is a biliprotein enriched in blue-green algae that is known to possess antioxidant, anti-apoptosis, anti-inflammatory, and radical-scavenging properties in somatic cells. However, the protective effect of CP on porcine embryo developmental competence in vitro remains unclear. In the present study, we investigated the effect of CP on the development of early porcine embryos as well as its underlying mechanisms. Different concentrations of CP (2, 5, 8, 10 µg/mL) were added to porcine zygote medium 5 during in vitro culture. The results showed that 5 µg/mL CP significantly increased blastocyst formation and hatching rate. Blastocyst formation and quality were significantly increased in the 50 µM H2O2 treatment group following 5 µg/mL CP addition. CP prevented the H2O2-induced compromise of mitochondrial membrane potential, release of cytochrome c from the mitochondria, and reactive oxygen species generation. Furthermore, apoptosis, DNA damage level, and autophagy in the blastocysts were attenuated by supplementation of CP in the H2O2-induced oxidative injury group compared to in controls. These results suggest that CP has beneficial effects on the development of porcine parthenotes by attenuating mitochondrial dysfunction and oxidative stress.


Subject(s)
Embryonic Development/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Parthenogenesis/drug effects , Phycocyanin/pharmacology , Protective Agents/pharmacology , Animals , Autophagy , Embryo Culture Techniques , Female , Hydrogen Peroxide/toxicity , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidants/toxicity , Reactive Oxygen Species/metabolism , Swine
18.
Cell Cycle ; 16(22): 2220-2229, 2017.
Article in English | MEDLINE | ID: mdl-28933982

ABSTRACT

Checkpoint 1 (Chk1), as an important member of DNA replication checkpoint and DNA damage response, has an important role during the G2/M stage of mitosis. In this study, we used porcine oocyte as a model to investigate the function of Chk1 during porcine oocyte maturation. Chk1 was expressed from germinal vesicle (GV) to metaphase II (MII) stages, mainly localized in the cytoplasm at GV stage and moved to the spindle after germinal vesicle breakdown (GVBD). Chk1 depletion not only induced oocytes to be arrested at MI stage with abnormal chromosomes arrangement, but also inhibited the degradation of Cyclin B1 and decreased the expression of Mitotic Arrest Deficient 2-Like 1 (Mad2L1), one of spindle assembly checkpoint (SAC) proteins, and cadherin 1 (Cdh1), one of coactivation for anaphase-promoting complex/cyclosome (APC/C). Moreover, Chk1 overexpression delayed GVBD. These results demonstrated that Chk1 facilitated the timely degradation of Cyclin B1 at anaphase I (AI) and maintained the expression of Mad2L1 and Cdh1, which ensured that all chromosomes were accurately located in a line, and then oocytes passed metaphase I (MI) and AI and exited from the first meiotic division successfully. In addition, we proved that Chk1 had not function on GVBD of porcine oocytes, which suggested that maturation of porcine oocytes did not need the DNA damage checkpoint, which was different from the mouse oocyte maturation.


Subject(s)
Checkpoint Kinase 1/metabolism , Oocytes/metabolism , Animals , Checkpoint Kinase 1/genetics , Meiosis/genetics , Meiosis/physiology , Metaphase/genetics , Metaphase/physiology , Swine
19.
Aging (Albany NY) ; 9(6): 1552-1564, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28657543

ABSTRACT

Oxidative stress is known as a major contributing factor involved in oocyte aging, which negatively affects oocyte quality and development after fertilization. Melatonin is an effective free radical scavenger and its metabolites AFMK and AMK are powerful detoxifiers that eliminate free radicals. In this study, we used porcine oocytes to test the hypothesis that melatonin could scavenge free radicals produced during oocyte aging, thereby maintaining oocyte quality. We compared reactive oxygen species levels, apoptosis levels, mitochondrial membrane potential ratios, total glutathione contents and expression levels in fresh, aged and melatonin-treated aged porcine oocytes and observed the percentage of blastocyst formation following parthenogenetic activation. We found that melatonin could effectively maintain the morphology of oocytes observed in control oocytes, alleviate oxidative stress, markedly decrease early apoptosis levels, retard the decline of mitochondrial membrane potential and significantly promote subsequent embryonic development in oocytes aged for 24 hr in vitro. These results strongly suggest that melatonin can prevent postovulatory oocyte aging and promote subsequent embryonic development in the pig, which might find practical applications to control oocyte aging in other mammalian species including humans to maintain the quality of human oocytes when performing clinical assisted reproductive technology.


Subject(s)
Embryo Culture Techniques/veterinary , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Melatonin/pharmacology , Oocytes/drug effects , Swine/embryology , Animals , Antioxidants , Gene Expression Regulation, Developmental/drug effects , Melatonin/administration & dosage , Melatonin/metabolism , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects
20.
Oncotarget ; 8(13): 20602-20611, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28206974

ABSTRACT

Caffeine, as an oocyte aging inhibitor, was used in many different species to control or delay oocyte aging. However, the safety of caffeine and developmental competence of aged oocytes inhibited by caffeine has not been studied systematically. So we detected the spindle morphology, distribution of cortical granules, zona pellucida hardening and pronucleus formation to assess oocyte quality of caffeine treated oocytes. We found that aged oocytes treated by caffeine maintained weak susceptibility to activating stimuli and regained normal competent after aged further 6 hr. Caffeine maintained the spindle morphology, changed cortical granules distribution of aged oocytes and could not prevent zona pellucida hardening. Furthermore, caffeine increased pronucleus formation of aged oocytes and decreased fragmentation after fertilization. These results suggested that caffeine could maintain the quality of aged oocytes safely in mouse.


Subject(s)
Caffeine/pharmacology , Cellular Senescence/drug effects , Oocytes/drug effects , Purinergic P1 Receptor Antagonists/pharmacology , Animals , Female , Fluorescent Antibody Technique , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...