Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Aging ; 3(11): 1372-1386, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37845508

ABSTRACT

Advanced age is a primary risk factor for female infertility due to reduced ovarian reserve and declining oocyte quality. However, as an important contributing factor, the role of metabolic regulation during reproductive aging is poorly understood. Here, we applied untargeted metabolomics to identify spermidine as a critical metabolite in ovaries to protect oocytes against aging. In particular, we found that the spermidine level was reduced in ovaries of aged mice and that supplementation with spermidine promoted follicle development, oocyte maturation, early embryonic development and female fertility of aged mice. By microtranscriptomic analysis, we further discovered that spermidine-induced recovery of oocyte quality was mediated by enhancement of mitophagy activity and mitochondrial function in aged mice, and this mechanism of action was conserved in porcine oocytes under oxidative stress. Altogether, our findings suggest that spermidine supplementation could represent a therapeutic strategy to ameliorate oocyte quality and reproductive outcome in cis-gender women and other persons trying to conceive at an advanced age. Future work is needed to test whether this approach can be safely and effectively translated to humans.


Subject(s)
Polyamines , Spermidine , Pregnancy , Female , Humans , Mice , Animals , Swine , Spermidine/pharmacology , Polyamines/metabolism , Mitophagy , Oocytes , Aging
2.
Ecotoxicol Environ Saf ; 254: 114768, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36917878

ABSTRACT

Diisononyl phthalate (DINP), a mixture of chemical compounds composed of diverse isononyl esters of phthalic acid, is commonly applied as a plasticizer to substitute for di (2-ethylhexyl) phthalate (DEHP). It has been demonstrated that DINP exposure impairs the functions of kidney and liver in animals. However, the effects and potential mechanisms of DINP exposure on the female reproduction, especially the oocyte quality are still poorly understood. Here, we discovered that DINP exposure weakened the porcine oocyte meiotic competency (78.9% vs 53.6%, P < 0.001) and fertilization ability (78.5% vs 34.1%, P < 0.0001) during in vitro maturation. Specifically, DINP exposure induced the persistent spindle assembly checkpoint (SAC) activation caused by the disorganized spindle/chromosome apparatus (spindle: 20.0% vs 83.3%, P < 0.001; chromosome: 20.0% vs 80.0%, P < 0.01) to arrest meiotic progression of oocytes at metaphase I stage. In addition, DINP exposure disturbed the dynamics of sperm binding (146.7 vs 58.6, P < 0.0001) and fusion proteins (19.5 vs 11.6, P < 0.0001) in oocytes to compromise their fertilization ability. In particular, transcriptome data uncovered that the action mechanism of DINP on the oocyte maturation was associated with oxidative phosphorylation, apoptosis and autophagy pathways. Lastly, we validated that DINP exposure resulted in the mitochondrial dysfunction (27.2 vs 19.8, P < 0.0001) and elevated levels of reactive oxygen species (ROS; 8.9 vs 19.9, P < 0.0001) to trigger the occurrence of apoptosis (7.2 vs 13.1, P < 0.0001) and protective autophagy (68.6 vs 139.3, P < 0.01). Altogether, our findings not only testify that DINP has a potentially adverse impact on the mammalian oocyte quality, but also provide a scientific reference regarding how environment pollutants act on the female germ cell development.


Subject(s)
Diethylhexyl Phthalate , Phthalic Acids , Male , Female , Swine , Animals , Diethylhexyl Phthalate/metabolism , Semen , Phthalic Acids/metabolism , Oocytes , Apoptosis , Mammals
4.
Genome Biol ; 23(1): 5, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980211

ABSTRACT

BACKGROUND: Direct analogs of chemically modified bases that carry important epigenetic information, such as 5-methylcytosine (m5C)/5-methyldeoxycytosine (5mC), 5-hydroxymethylcytosine (hm5C)/5-hydroxymethyldeoxycytosine (5hmC), and N6-methyladenosine (m6A)/N6-methyldeoxyadenosine (6mA), are detected in both RNA and DNA, respectively. The modified base N4-acetylcytosine (ac4C) is well studied in RNAs, but its presence and epigenetic roles in cellular DNA have not been explored. RESULTS: Here, we demonstrate the existence of N4-acetyldeoxycytosine (4acC) in genomic DNA of Arabidopsis with multiple detection methods. Genome-wide profiling of 4acC modification reveals that 4acC peaks are mostly distributed in euchromatin regions and present in nearly half of the expressed protein-coding genes in Arabidopsis. 4acC is mainly located around transcription start sites and positively correlates with gene expression levels. Imbalance of 5mC does not directly affect 4acC modification. We also characterize the associations of 4acC with 5mC and histone modifications that cooperatively regulate gene expression. Moreover, 4acC is also detected in genomic DNA of rice, maize, mouse, and human by mass spectrometry. CONCLUSIONS: Our findings reveal 4acC as a hitherto unknown DNA modification in higher eukaryotes. We identify potential interactions of this mark with other epigenetic marks in gene expression regulation.


Subject(s)
Arabidopsis , 5-Methylcytosine , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Cytosine/metabolism , DNA/metabolism , DNA Methylation , Epigenomics , Euchromatin , Mice
5.
Cell Death Differ ; 29(8): 1466-1473, 2022 08.
Article in English | MEDLINE | ID: mdl-35017645

ABSTRACT

Soon after fertilization, the block mechanisms are developed in the zona pellucida (ZP) and plasma membrane of the egg to prevent any additional sperm from binding, penetration, and fusion. However, the molecular basis and underlying mechanism for the post-fertilization block to sperm penetration through ZP has not yet been determined. Here, we find that transglutaminase 2 (Tgm2), an enzyme that catalyzes proteins by the formation of an isopeptide bond within or between polypeptide chains, crosslinks zona pellucida glycoprotein 3 (ZP3) to result in the ZP hardening after fertilization and thus prevents polyspermy. Tgm2 abundantly accumulates in the subcortical region of the oocytes and vanishes upon fertilization. Both inhibition of Tgm2 activity in oocytes by the specific inhibitor in vitro and genetic ablation of Tgm2 in vivo cause the presence of additional sperm in the perivitelline space of fertilized eggs, consequently leading to the polyploid embryos. Biochemically, recombinant Tgm2 binds to and crosslinks ZP3 proteins in vitro, and incubation of oocytes with recombinant Tgm2 protein inhibits the polyspermy. Altogether, our data identify Tgm2 as a participant of zona block to the post-fertilization sperm penetration via hardening ZP surrounding fertilized eggs, extending our current understanding about the molecular basis of block to polyspermy.


Subject(s)
Protein Glutamine gamma Glutamyltransferase 2 , Semen , Zona Pellucida Glycoproteins , Animals , Female , Male , Mice , Oocytes , Protein Glutamine gamma Glutamyltransferase 2/genetics , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Proteins/metabolism , Sperm-Ovum Interactions , Spermatozoa/metabolism , Zona Pellucida/chemistry , Zona Pellucida/metabolism , Zona Pellucida Glycoproteins/genetics , Zona Pellucida Glycoproteins/metabolism
6.
Environ Pollut ; 284: 117508, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34261219

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a widely used artificial surfactant with potential toxicity to humans and animals. However, little is known about the impact of PFOS on the female germ cell development. Here, we report that PFOS exposure weakens oocyte quality by disturbing oocyte meiotic competency and fertilization ability. Specifically, PFOS exposure impaired cytoskeleton assembly including spindle organization and actin polymerization to cause the oocyte maturation arrest. In addition, PFOS exposure also impaired the mitochondrial dynamics and function, resulting in the increased levels of reactive oxygen species (ROS) and DNA damage as well as generation of apoptosis. Lastly, PFOS exposure compromised the distribution of cortical granules (CGs) and their component ovastacin, leading to the failure of sperm binding and fertilization. Altogether, our study illustrates that oxidative stress-induced apoptosis is a major cause for the deteriorated quality of porcine oocytes exposed to PFOS.


Subject(s)
Alkanesulfonic Acids , Meiosis , Alkanesulfonic Acids/metabolism , Alkanesulfonic Acids/toxicity , Animals , Apoptosis , Female , Fluorocarbons , Humans , Oocytes/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Swine
7.
Front Cell Dev Biol ; 9: 678665, 2021.
Article in English | MEDLINE | ID: mdl-34150773

ABSTRACT

Copper (Cu) is an essential trace element for animals, and also an important nutritional component for the normal physiology and metabolism of animal reproductive systems. An excess or lack of Cu will directly or indirectly affect animal reproductive activities. However, the effect of Cu, in particular excessive Cu, on the reproductive performance of sows has not been studied. Here, we report that excessive Cu had negative effects on oocyte maturation and organelle functions. We showed that Cu exposure perturbed porcine oocyte meiotic maturation and impaired spindle/chromosome structure, resulting in a defective spindle assembly, as well as the abnormal distribution of actin dynamics and cortical granules. In addition, single-cell transcriptome analysis identified the target effectors of Cu actions in porcine oocytes, further demonstrating that Cu exposure affects the mitochondrial distribution and function, leading to the high levels of reactive oxygen species, DNA damage, and early apoptosis of porcine oocytes. These findings demonstrate that Cu exposure causes abnormalities in the mitochondrial distribution and function, resulting in the increased oxidative stress and levels of reactive oxygen species, DNA damage, and apoptosis, ultimately leading to a decreased porcine oocyte quality.

8.
Cell Rep ; 32(5): 107987, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755581

ABSTRACT

Advanced maternal age is highly associated with a decline in oocyte quality, but effective approaches to improve it have still not been fully determined. Here, we report that in vivo supplementation of nicotinamide mononucleotide (NMN) efficaciously improves the quality of oocytes from naturally aged mice by recovering nicotinamide adenine dinucleotide (NAD+) levels. NMN supplementation not only increases ovulation of aged oocytes but also enhances their meiotic competency and fertilization ability by maintaining the normal spindle/chromosome structure and the dynamics of the cortical granule component ovastacin. Moreover, single-cell transcriptome analysis shows that the beneficial effect of NMN on aged oocytes is mediated by restoration of mitochondrial function, eliminating the accumulated ROS to suppress apoptosis. Collectively, our data reveal that NMN supplementation is a feasible approach to protect oocytes from advanced maternal age-related deterioration, contributing to the improvement of reproductive outcome of aged women and assisted reproductive technology.


Subject(s)
Aging/physiology , Cellular Senescence , Nicotinamide Mononucleotide/pharmacology , Oocytes/cytology , Animals , Apoptosis/drug effects , Cellular Senescence/drug effects , Chromosomes, Mammalian/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , DNA Damage , Dietary Supplements , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Fertilization/drug effects , Kinetochores/drug effects , Kinetochores/metabolism , Male , Meiosis/drug effects , Metalloproteases/metabolism , Mice, Inbred ICR , Microtubules/drug effects , Microtubules/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Oocytes/drug effects , Reactive Oxygen Species/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Transcriptome/genetics
9.
Sci Adv ; 6(15): eaax3969, 2020 04.
Article in English | MEDLINE | ID: mdl-32284991

ABSTRACT

During mitotic prophase, cohesins are removed from chromosome arms by Wapl to ensure faithful sister chromatid separation. However, during female meiosis I, the resolution of chiasmata requires the proteolytic cleavage of cohesin subunit Rec8 along chromosome arms by Separase to separate homologs, and thus the role of Wapl remained unknown. Here, we report that Wapl functions as a regulator of spindle assembly checkpoint (SAC) to prevent aneuploidy in meiosis I. Depletion of Wapl accelerates meiotic progression, inactivates SAC, and causes meiotic defects such as aberrant spindle/chromosome structure and incorrect kinetochore-microtubule (K-MT) attachment, consequently leading to aneuploid eggs. Notably, we identify Bub3 as a binding partner of Wapl by immunoprecipitation and mass spectrometry analysis. We further determine that Wapl controls the SAC activity by maintaining Bub3 protein level and document that exogenous Bub3 restores the normal meiosis in Wapl-depleted oocytes. Together, our findings uncover unique, noncanonical roles for Wapl in mediating control of the SAC in female meiosis I.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , M Phase Cell Cycle Checkpoints , Meiosis , Poly-ADP-Ribose Binding Proteins/metabolism , Proteins/metabolism , Aneuploidy , Animals , Chromosome Pairing , Female , Mice , Models, Biological , Oocytes/metabolism
10.
J Anim Sci Biotechnol ; 11: 31, 2020.
Article in English | MEDLINE | ID: mdl-32292585

ABSTRACT

BACKGROUND: CK2 (casein kinase 2) is a serine/threonine-selective protein kinase that has been involved in a variety of cellular processes such as DNA repair, cell cycle control and circadian rhythm regulation. However, its functional roles in oocyte meiosis have not been fully determined. RESULTS: We report that CK2 is essential for porcine oocyte meiotic maturation by regulating spindle assembly checkpoint (SAC). Immunostaining and immunoblotting analysis showed that CK2 was constantly expressed and located on the chromosomes during the entire oocyte meiotic maturation. Inhibition of CK2 activity by its selective inhibitor CX-4945 impaired the first polar body extrusion and arrested oocytes at M I stage, accompanied by the presence of BubR1 at kinetochores, indicative of activated SAC. In addition, we found that spindle/chromosome structure was disrupted in CK2-inhibited oocytes due to the weakened microtubule stability, which is a major cause resulting in the activation of SAC. Last, we found that the level DNA damage as assessed by γH2A.X staining was considerably elevated when CK2 was inhibited, suggesting that DNA damage might be another critical factor leading to the SAC activation and meiotic failure of oocytes. CONCLUSIONS: Our findings demonstrate that CK2 promotes the porcine oocyte maturation by ensuring normal spindle assembly and DNA damage repair.

11.
Cell Cycle ; 18(19): 2481-2494, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31387516

ABSTRACT

In mammalian cells, cohesin acetyltransferases Esco1 and Esco2 acetylate cohesin subunit Smc3 to establish chromosome cohesion, ensuring the accurate chromosome segregation. However, we have previously documented that both Esco1 and Esco2 have unique substrates and roles in mouse oocyte meiosis I to orchestrate the meiotic progression, but whether these functions are conserved among species is still not determined. Here, we used porcine oocytes as a model to illustrate that Esco1 and Esco2 exerted conserved functions during oocyte meiosis. We observed that Esco1 and Esco2 exhibited different localization patterns in porcine oocytes. Esco1 was localized to the spindle apparatus while Esco2 was distributed on the chromosomes. Depletion of Esco1 by siRNA microinjection caused the meiotic arrest by showing the reduced frequency of first polar body extrusion and defective spindle/chromosome structure. In addition, Esco1 bound to α-tubulin and was required for its acetylation level to maintain the microtubule dynamics. By contrast, depletion of Esco2 by siRNA microinjection resulted in the accelerated meiotic progression by displaying the precocious polar body extrusion and inactivation of spindle assembly checkpoint. Notably, Esco2 was shown to be associated with histone H4 for the acetylation of H4K16 to modulate the kinetochore function. Collectively, our data reveal that Esco1 and Esco2 perform distinct and conserved functions in oocytes to drive the meiotic progression beyond their canonical roles in the cohesion establishment.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Meiosis/genetics , Oocytes/metabolism , Spindle Apparatus/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Chromatids/metabolism , Chromosome Segregation/genetics , Gene Knockdown Techniques , Histones/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/genetics , RNA, Small Interfering , Swine , Tubulin/metabolism , Cohesins
12.
Free Radic Biol Med ; 143: 84-94, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31398498

ABSTRACT

Postovulatory aging is known to compromise the oocyte quality as well as subsequent embryo development in many different animal models, and becomes one of the most intractable issues that limit the outcome of human assisted reproductive technology (ART). However, the strategies to prevent the deterioration of aged oocytes and relevant mechanisms are still underexplored. Here, we find that supplementation of CoQ10, a natural antioxidant present in human follicular fluids, is able to restore the postovulatory aging-induced fragmentation of oocytes and decline of fertilization. Importantly, we show that CoQ10 supplementation recovers postovulatory aging-caused meiotic defects such as disruption of spindle assembly, misalignment of chromosome, disappearance of actin cap, and abnormal distribution patterns of mitochondria and cortical granules. In addition, CoQ10 protects aged oocytes from premature exocytosis of ovastacin, cleavage of sperm binding site ZP2, and loss of localization of Juno, to maintain the fertilization potential. Notably, CoQ10 suppresses the aging-induced oxidative stress by reducing the levels of superoxide and DNA damage, ultimately inhibiting the apoptosis. Taken together, our findings demonstrate that CoQ10 supplementation is a feasible and effective way to prevent postovulatory aging and preserve the oocyte quality, potentially contributing to improve the successful rate of IVF (in vitro fertilization) and ICSI (intracytoplasmic sperm injection) during human ART.


Subject(s)
Apoptosis , Cellular Senescence , DNA Damage , Oocytes/drug effects , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Chromosomes/metabolism , Female , Fertilization in Vitro , Free Radical Scavengers , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred ICR , Oxidative Stress , Reproductive Techniques, Assisted , Sperm Injections, Intracytoplasmic , Superoxides/metabolism , Ubiquinone/pharmacology
13.
Cell Death Dis ; 10(7): 474, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31209201

ABSTRACT

Reproductive problem has been one of the top issues for women health worldwide in recent decades. As a typical female disease, primary ovarian insufficiency (POI) results in a loss of ovarian follicles and oocytes that thus destroys women fertility. However, due to the complex of POI etiology and rare resource of human POI oocytes, few biomarkers have been identified in clinics and no effective strategy could be applied to treat POI patients. In the search of possible association between DNA damage and POI by Smart-Seq2 and RT2 profiler PCR array, we find that BRCA2, a core DNA repair gene for homologous recombination shows significantly lower expression in two POI patient oocytes. In line with this, we generated oocyte-specific knockout mouse model driven by Gdf9-Cre. The Brca2-deficient mice are infertile because of the arrested follicle development and defective oocyte quality caused by the accumulation of DNA damage. Notably, ectopic expression of Brca2 in Brca2-deficient oocytes could partially restore the oocyte maturation and chromosome stability. Collectively, our data assign a definite deficiency to BRCA2 as a POI driver during follicle development and oocyte maturation, and provide a potential fertility treatment strategy for POI patients induced by BRCA2 deficiency.


Subject(s)
BRCA2 Protein/deficiency , Primary Ovarian Insufficiency/metabolism , Alleles , Animals , BRCA2 Protein/metabolism , DNA Damage/genetics , Down-Regulation/genetics , Exons/genetics , Female , Fertility , Gene Deletion , Humans , Meiosis , Mice , Oocytes/metabolism , Ovarian Follicle/metabolism , Primary Ovarian Insufficiency/genetics
14.
J Cell Physiol ; 234(11): 20111-20117, 2019 11.
Article in English | MEDLINE | ID: mdl-30950061

ABSTRACT

Brefeldin A (BFA) is a lactone antibiotic synthesized from palmitic acid by several fungi that could block anterograde transport of proteins from endoplasmic reticulum to Golgi apparatus by reversible disruption of the Golgi complex. Previous investigations have shown that BFA induces the apoptosis of cancer cells in mitosis and impairs asymmetric spindle positioning in meiosis. Here, we document that exposure to BFA in porcine oocytes compromises the meiotic maturation via disrupting both nuclear and cytoplasmic maturation. We found that BFA exposure collapsed the cytoskeleton assembly by showing the aberrant spindle organization with misaligned chromosomes and defective actin dynamics. Furthermore, the distribution of both mitochondria and cortical granules (CGs), two important indexes of cytoplasmic maturation of oocytes, was disturbed following BFA exposure. We finally validated that the localization of ovastacin, a component of CGs that is essential for the postfertilization removal of sperm-binding sites in the zona pellucida, was also perturbed in BFA-exposed oocytes, which might weaken their fertilization capacity. Collectively, these findings indicate that Golgi-mediated protein transport is indispensable for the porcine oocyte meiotic maturation.


Subject(s)
Brefeldin A/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Actins/metabolism , Animals , Apoptosis/drug effects , Chromosomes/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Fertilization/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Microtubules/drug effects , Microtubules/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitosis/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Swine , Zona Pellucida/drug effects , Zona Pellucida/metabolism
15.
Nucleic Acids Res ; 46(5): 2335-2346, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29361031

ABSTRACT

Esco1 has been reported to function as a cohesion establishment factor that mediates chromosome cohesion and segregation in mitotic cells. However, its exact roles in meiosis have not been clearly defined. Here, we document that Esco1 is expressed and localized to both the nucleus and cytoplasm during mouse oocyte meiotic maturation. Depletion of Esco1 by siRNA microinjection causes the meiotic progression arrest with a severe spindle abnormality and chromosome misalignment, which is coupled with a higher incidence of the erroneous kinetochore-microtubule attachments and activation of spindle assembly checkpoint. In addition, depletion of Esco1 leads to the impaired microtubule stability shown by the weakened resistance ability to the microtubule depolymerizing drug nocodazole and the decreased level of acetylated α-tubulin. Conversely, overexpression of Esco1 causes hyperacetylation of α-tubulin and spindle defects. Moreover, we find that Esco1 binds to α-tubulin and is required for its acetylation. The reduced acetylation level of α-tubulin in Esco1-depleted oocytes can be restored by the ectopic expression of exogenous wild-type Esco1 but not enzymatically dead Esco1-G768D. Purified wild-type Esco1 instead of mutant Esco1-G768D acetylates the synthesized peptide of α-tubulin in vitro. Collectively, our data assign a novel function to Esco1 as a microtubule regulator during oocyte meiotic maturation beyond its conventional role in chromosome cohesion.


Subject(s)
Acetyltransferases/metabolism , Meiosis , Oocytes/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism , Acetylation , Acetyltransferases/physiology , Animals , Chromosomes, Mammalian , Cytoplasm/metabolism , Female , Kinetochores/metabolism , Lysine/metabolism , M Phase Cell Cycle Checkpoints , Meiosis/genetics , Mice, Inbred ICR , Microtubules/metabolism , Oocytes/enzymology , Tubulin/chemistry
16.
FASEB J ; 32(3): 1328-1337, 2018 03.
Article in English | MEDLINE | ID: mdl-29109171

ABSTRACT

Negative effects of postovulatory aging on fertilization ability and subsequent embryo development have been reported in rodents; however, the molecular and cellular changes during this process have not been fully defined. Here, we used porcine oocytes, a model that is physiologically and developmentally similar to humans, to explore the molecular mechanisms that underlie how postovulatory aging affects oocyte quality and fertilization capacity. We found that postovulatory aging caused the morphologic change of porcine oocytes by exhibiting the incompact expansion of cumulus cells and an increased occurrence of fragmentation. Aging also impaired oocyte quality by disrupting organelle structures, including the spindle assembly, actin polymerization, and mitochondrial integrity. Moreover, postovulatory aging led to the abnormal distribution of cortical granules and ovastacin, which, in turn, resulted in defective sperm binding and consequently compromised fertilization potential. Of note, we observed that postovulatory aging induced oxidative stress with a high level of reactive oxygen species and apoptotic rate in oocytes, thereby resulting in the deterioration of critical factors in the maintenance of oocyte quality and fertilization capacity. Taken together, our findings demonstrate that postovulatory aging perturbs a variety of molecular and cellular changes in porcine oocytes by inducing oxidative stress.-Miao, Y., Zhou, C., Cui, Z., Zhang, M., ShiYang, X., Lu, Y., Xiong, B. Postovulatory aging causes the deterioration of porcine oocytes via induction of oxidative stress.


Subject(s)
Apoptosis , Cellular Senescence , Oocytes/pathology , Ovulation , Oxidative Stress , Spermatozoa/pathology , Animals , Cells, Cultured , Female , Male , Oocytes/metabolism , Reactive Oxygen Species/metabolism , Spermatozoa/metabolism , Swine
17.
Hum Reprod ; 33(1): 116-127, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29112712

ABSTRACT

STUDY QUESTION: Does melatonin restore the benzo(a)pyrene (BaP)-induced meiotic failure in porcine oocytes? SUMMARY ANSWER: Melatonin effectively inhibits the increased reactive oxygen species (ROS) level and apoptotic rate in BaP-exposed porcine oocytes to recover the meiotic failure. WHAT IS KNOWN ALREADY: BaP, a widespread environmental carcinogen found in particulate matter, 2.5 µm or less (PM2.5), has been shown to have toxicity at the level of the reproductive systems. BaP exposure disrupts the steroid balance, alters the expression of ovarian estrogen receptor and causes premature ovarian failure through the rapid depletion of the primordial follicle pool. In addition, acute exposure to BaP has transient adverse effects on the follicle growth, ovulation and formation of corpora lutea, which results in transient infertility. STUDY DESIGN, SIZE, DURATION: Porcine oocytes were randomly assigned to control, BaP-exposed and melatonin-supplemented groups. BaP was dissolved in dimethylsulphoxide and diluted to a final concentration of 50, 100 or 250 µM with maturation medium, respectively. Melatonin was dissolved in the absolute ethanol and diluted with maturation medium to a final concentration of 1 nM, 100 nM, 10 µM and 1 mM, respectively. The in vitro cultured oocytes from each group after treatment were applied to the subsequent analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: Acquisition of oocyte meiotic competence was assessed using immunostaining, fluorescent intensity quantification and/or immunoblotting to analyse the cytoskeleton assembly, mitochondrial integrity, cortical granule dynamics, ovastacin distribution, ROS level and apoptotic rate. Fertilization ability of oocytes was examined by sperm binding assay and IVF. MAIN RESULTS AND THE ROLE OF CHANCE: BaP exposure resulted in the oocyte meiotic failure (P = 0.001) via impairing the meiotic apparatus, showing a prominently defective spindle assembly (P = 0.003), actin dynamics (P < 0.001) and mitochondrion integrity (P < 0.001). In addition, BaP exposure caused the abnormal distribution of cortical granules (P < 0.001) and ovastacin (P = 0.003), which were consistent with the observation that fewer sperm bound to the zona pellucida surrounding the unfertilized BaP-exposed eggs (P < 0.001), contributing to the fertilization failure (P < 0.001). Conversely, melatonin supplementation recovered, at least partially, all the meiotic defects caused by BaP exposure through inhibiting the rise in ROS level (P = 0.015) and apoptotic rate (P = 0.001). LIMITATIONS, REASONS FOR CAUTION: We investigated the negative impact of BaP on the oocyte meiotic maturation in vitro, but not in vivo. WIDER IMPLICATIONS OF THE FINDINGS: Our findings not only deeply clarify the potential mechanisms of BaP-induced oocyte meiotic failure, but also extend the understanding about how environmental pollutants influence the reproductive systems in humans. STUDY FUNDING/COMPETING INTERESTS: This study was supported by the National Natural Science Foundation of China (31571545) and the Natural Science Foundation of Jiangsu Province (BK20150677). The authors have no conflict of interest to disclose.


Subject(s)
Benzo(a)pyrene/toxicity , Meiosis/drug effects , Melatonin/pharmacology , Oocytes/cytology , Oocytes/drug effects , Animals , Apoptosis/drug effects , Carcinogens, Environmental/toxicity , China , Female , Fertilization/drug effects , Humans , In Vitro Techniques , Male , Mitochondria/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Particulate Matter/toxicity , Reactive Oxygen Species/metabolism , Sperm-Ovum Interactions/drug effects , Sus scrofa
18.
FASEB J ; 32(1): 342-352, 2018 01.
Article in English | MEDLINE | ID: mdl-28904021

ABSTRACT

Benzo[a]pyrene (BaP) is a ubiquitous environmental pollutant and carcinogen that is frequently found in particulate matter, with a diameter of ≤2.5 µm (PM2.5). It has been reported to interrupt the normal reproductive system, but the exact molecular basis has not been clearly defined. To understand the underlying mechanisms regarding how BaP exposure disrupts female fertility, we evaluated oocyte quality by assessing the critical regulators and events during oocyte meiotic maturation and fertilization. We found that BaP exposure compromised the mouse oocyte meiotic progression by disrupting normal spindle assembly, chromosome alignment, and kinetochore-microtubule attachment, consequently leading to the generation of aneuploid eggs. In addition, BaP administration significantly decreased the fertilization rate of mouse eggs by reducing the number of sperm binding to the zona pellucida, which was consistent with the premature cleavage of N terminus of zona pellucida sperm-binding protein 2 and precocious exocytosis of ovastacin. Furthermore, BaP exposure interfered with the gamete fusion process by perturbing the localization and protein level of Juno. Notably, we found that BaP exposure induced oxidative stress with an increased level of reactive oxygen species and apoptosis in oocytes and thereby led to the deterioration of critical regulators and events during oocyte meiotic progression and fertilization. Our data document that BaP exposure reduces female fertility via impairing oocyte maturation and fertilization ability induced by oxidative stress and early apoptosis in murine models.-Zhang, M., Miao, Y., Chen, Q., Cai, M., Dong, W., Dai, X., Lu, Y., Zhou, C., Cui, Z., Xiong, B. BaP exposure causes oocyte meiotic arrest and fertilization failure to weaken female fertility.


Subject(s)
Benzo(a)pyrene/toxicity , Fertilization/drug effects , Infertility, Female/chemically induced , Oocytes/drug effects , Oocytes/pathology , Aneugens/toxicity , Animals , Apoptosis/drug effects , Environmental Pollutants/toxicity , Female , Infertility, Female/pathology , Kinetochores/drug effects , Male , Meiosis/drug effects , Mice , Mice, Inbred ICR , Microtubules/drug effects , Oocytes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sperm-Ovum Interactions/drug effects
19.
Cell Cycle ; 16(21): 2139-2145, 2017.
Article in English | MEDLINE | ID: mdl-28933593

ABSTRACT

Cytoplasmic dynein is a family of cytoskeletal motor proteins that move towards the minus-end of the microtubules to perform functions in a variety of mitotic processes such as cargo transport, organelle positioning, chromosome movement and centrosome assembly. However, its specific roles during mammalian oocyte meiosis have not been fully defined. Herein, we investigated the critical events during porcine oocyte meiotic maturation after inhibition of dynein by Ciliobrevin D treatment. We found that oocyte meiotic progression was arrested when inhibited of dynein by showing the poor expansion of cumulus cells and decreased rate of polar body extrusion. Meanwhile, the spindle assembly and chromosome alignment were disrupted, accompanied by the reduced level of acetylated α-tubulin, indicative of weakened microtubule stability. Defective actin polymerization on the plasma membrane was also observed in dynein-inhibited oocytes. In addition, inhibition of dynein caused the abnormal distribution of cortical granules and precocious exocytosis of ovastacin, a cortical granule component, which predicts that ZP2, the sperm binding site in the zona pellucida, might be prematurely cleaved in the unfertilized dynein-inhibited oocytes, potentially leading to the fertilization failure. Collectively, our findings reveal that dynein plays a part in porcine oocyte meiotic progression by regulating the cytoskeleton dynamics including microtubule stability, spindle assembly, chromosome alignment and actin polymerization. We also find that dynein mediates the normal cortical granule distribution and exocytosis timing of ovastacin in unfertilized eggs which are the essential for the successful fertilization.


Subject(s)
Cytoskeleton/metabolism , Dyneins/metabolism , Oocytes/metabolism , Animals , Centrosome/metabolism , Chromosomes/metabolism , Cumulus Cells/metabolism , Meiosis/physiology , Oogenesis/physiology , Swine
20.
Nucleic Acids Res ; 45(16): 9388-9397, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934466

ABSTRACT

Sister chromatid cohesion, mediated by cohesin complex and established by the acetyltransferases Esco1 and Esco2, is essential for faithful chromosome segregation. Mutations in Esco2 cause Roberts syndrome, a developmental disease characterized by severe prenatal retardation as well as limb and facial abnormalities. However, its exact roles during oocyte meiosis have not clearly defined. Here, we report that Esco2 localizes to the chromosomes during oocyte meiotic maturation. Depletion of Esco2 by morpholino microinjection leads to the precocious polar body extrusion, the escape of metaphase I arrest induced by nocodazole treatment and the loss of BubR1 from kinetochores, indicative of inactivated SAC. Furthermore, depletion of Esco2 causes a severely impaired spindle assembly and chromosome alignment, accompanied by the remarkably elevated incidence of defective kinetochore-microtubule attachments which consequently lead to the generation of aneuploid eggs. Notably, we find that the involvement of Esco2 in SAC and kinetochore functions is mediated by its binding to histone H4 and acetylation of H4K16 both in vivo and in vitro. Thus, our data assign a novel meiotic function to Esco2 beyond its role in the cohesion establishment during mouse oocyte meiosis.


Subject(s)
Acetyltransferases/metabolism , Histones/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/genetics , Meiosis/genetics , Oocytes/enzymology , Acetylation , Acetyltransferases/physiology , Aneuploidy , Animals , Chromosomes, Mammalian/enzymology , Female , Histones/chemistry , Lysine/metabolism , Mice, Inbred ICR , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...