Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.008
Filter
1.
PeerJ ; 12: e17864, 2024.
Article in English | MEDLINE | ID: mdl-39221285

ABSTRACT

Meiosis is a critical process in sexual reproduction, and errors during this cell division can significantly impact fertility. Successful meiosis relies on the coordinated action of numerous genes involved in DNA replication, strand breaks, and subsequent rejoining. DNA topoisomerase enzymes play a vital role by regulating DNA topology, alleviating tension during replication and transcription. To elucidate the specific function of DNA topoisomerase 1α ( A t T O P 1 α ) in male reproductive development of Arabidopsis thaliana, we investigated meiotic cell division in Arabidopsis flower buds. Combining cytological and biochemical techniques, we aimed to reveal the novel contribution of A t T O P 1 α to meiosis. Our results demonstrate that the absence of A t T O P 1 α leads to aberrant chromatin behavior during meiotic division. Specifically, the top1α1 mutant displayed altered heterochromatin distribution and clustered centromere signals at early meiotic stages. Additionally, this mutant exhibited disruptions in the distribution of 45s rDNA signals and a reduced frequency of chiasma formation during metaphase I, a crucial stage for genetic exchange. Furthermore, the atm-2×top1α1 double mutant displayed even more severe meiotic defects, including incomplete synapsis, DNA fragmentation, and the presence of polyads. These observations collectively suggest that A t T O P 1 α plays a critical role in ensuring accurate meiotic progression, promoting homologous chromosome crossover formation, and potentially functioning in a shared DNA repair pathway with ATAXIA TELANGIECTASIA MUTATED (ATM) in Arabidopsis microspore mother cells.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Chromosome Segregation , DNA Topoisomerases, Type I , Meiosis , Arabidopsis/genetics , Arabidopsis/enzymology , Meiosis/physiology , Meiosis/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Recombination, Genetic , Mutation
2.
Zool Res ; 45(5): 1037-1047, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39147718

ABSTRACT

Serine protease 50 (PRSS50/TSP50) is highly expressed in spermatocytes. Our study investigated its role in testicular development and spermatogenesis. Initially, PRSS50 knockdown was observed to impair DNA synthesis in spermatocytes. To further explore this, we generated PRSS50 knockout ( Prss50 -/- ) mice ( Mus musculus), which exhibited abnormal spermatid nuclear compression and reduced male fertility. Furthermore, dysplastic seminiferous tubules and decreased sex hormones were observed in 4-week-old Prss50 -/- mice, accompanied by meiotic progression defects and increased apoptosis of spermatogenic cells. Mechanistic analysis indicated that PRSS50 deletion resulted in increased phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) and elevated levels of MAP kinase phosphatase 3 (MKP3), a specific ERK antagonist, potentially accounting for testicular dysplasia in adolescent Prss50 -/- mice. Taken together, these findings suggest that PRSS50 plays an important role in testicular development and spermatogenesis, with the MKP3/ERK signaling pathway playing a significant role in this process.


Subject(s)
MAP Kinase Signaling System , Meiosis , Mice, Knockout , Spermatozoa , Animals , Male , Mice , Meiosis/physiology , Spermatozoa/physiology , Spermatogenesis/physiology , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Testis/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
3.
Reprod Biol Endocrinol ; 22(1): 110, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198846

ABSTRACT

The ubiquitination is crucial for controlling cellular homeostasis and protein modification, in which ubiquitin-conjugating enzyme (E2) acts as the central player in the ubiquitination system. Ubiquitin-conjugating enzymes, which have special domains that catalyse substrates, have sequence discrepancies and modulate various pathophysiological processes in different cells of multiple organisms. E2s take part in the mitosis of primordial germ cells, meiosis of spermatocytes and the formation of mature haploid spermatids to maintain normal male fertility. In this review, we summarize the various types of E2s and their functions during distinct stages of spermatogenesis.


Subject(s)
Spermatogenesis , Ubiquitin-Conjugating Enzymes , Spermatogenesis/physiology , Male , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/physiology , Animals , Ubiquitination/physiology , Meiosis/physiology
4.
Theriogenology ; 229: 16-22, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39142066

ABSTRACT

Spermatogenesis in eukaryotes is a process that occurs within a very narrow temperature threshold, typically not exceeding 36 °C. SPO11 was isolated from the temperature-sensitive mutant receptor of Saccharomyces cerevisiae and is thought to be the only protein that functions during meiosis. This suggested that SPO11 may be the key protein that influenced the temperature of spermatogenesis not exceeding 36 °C. Elevated temperatures typically damage the spermatogenic cells. Birds have a core body temperature of 41-42 °C, and their testis are located inside their bodies, providing an alternative perspective to investigate the potential impact of temperature threshold on spermatogenesis. The objective of this study was to ascertain whether elevated ambient temperatures affect spermatogenesis in birds and whether SPO11 is the key gene affecting the temperature threshold for spermatogenesis. STRA8, SCP3, SPO11, γ-H2AX, and RAD51 were all crucial components in the process of meiotic initiation, synapsis, DNA double-strand break (DSB) induction, homologous chromosome crossover recombination, and repair of DSB. In this study, 39-day-old Japanese quail were subjected to heat stress (HS) at 38 °C for 8 h per day for 3 (3d HS) and 13 (13d HS) consecutive days and analyzed the expression of meiotic signaling molecules (STRA8, SCP3, SPO11, γ-H2AX, and RAD51) using molecular biology techniques, including Immunohistochemistry (IHC), Western Blot (WB), and Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). We found that spermatogenesis was normal in both groups exposed to HS. Meiotic signaling molecules were expressed normally in the 3d HS group. All detected signaling molecules were normally expressed in the 13d HS group, except for SPO11, which showed a significant increase in expression, indicating that SPO11 was temperature-sensitive. We examined the localized expression of each meiotic signaling molecule in quail testis, explored the temperature sensitivity of SPO11, and determined that quail testis can undergo normal spermatogenesis at ambient temperatures exceeding 36 °C. This study concluded that SPO11 is not the key protein influencing spermatogenesis in birds. These findings enhance our understanding of avian spermatogenesis.


Subject(s)
Spermatogenesis , Testis , Animals , Male , Spermatogenesis/physiology , Testis/metabolism , Hot Temperature , Meiotic Prophase I/physiology , Coturnix/genetics , Coturnix/physiology , Coturnix/metabolism , Signal Transduction , Gene Expression Regulation/physiology , Meiosis/physiology
5.
Theriogenology ; 229: 147-157, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39178616

ABSTRACT

Calcium ions (Ca2+) regulate cell proliferation and differentiation and participate in various physiological activities of cells. The calcium transfer protein inositol 1,4,5-triphosphate receptor (IP3R), located between the endoplasmic reticulum (ER) and mitochondria, plays an important role in regulating Ca2+ levels. However, the mechanism by which IP3R1 affects porcine meiotic progression and embryonic development remains unclear. We established a model in porcine oocytes using siRNA-mediated knockdown of IP3R1 to investigate the effects of IP3R1 on porcine oocyte meiotic progression and embryonic development. The results indicated that a decrease in IP3R1 expression significantly enhanced the interaction between the ER and mitochondria. Additionally, the interaction between the ER and the mitochondrial Ca2+ ([Ca2+]m) transport network protein IP3R1-GRP75-VDAC1 was disrupted. The results of the Duolink II in situ proximity ligation assay (PLA) revealed a weakened pairwise interaction between IP3R1-GRP75 and VDAC1 and a significantly increased interaction between GRP75 and VDAC1 after IP3R1 interference, resulting in the accumulation of large amounts of [Ca2+]m. These changes led to mitochondrial oxidative stress, increased the levels of reactive oxygen species (ROS) and reduced ATP production, which hindered the maturation and late development of porcine oocytes and induced apoptosis. Nevertheless, after treat with [Ca2+]m chelating agent ruthenium red (RR) or ROS scavenger N-acetylcysteine (NAC), the oocytes developmental abnormalities, oxidative stress and apoptosis caused by Ca2+ overload were improved. In conclusion, our results indicated IP3R1 is required for meiotic progression and embryonic development by regulating mitochondrial calcium and oxidative damage.


Subject(s)
Calcium , Embryonic Development , Inositol 1,4,5-Trisphosphate Receptors , Meiosis , Mitochondria , Oxidative Stress , Animals , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Swine , Mitochondria/metabolism , Mitochondria/physiology , Meiosis/physiology , Calcium/metabolism , Embryonic Development/physiology , Reactive Oxygen Species/metabolism , Oocytes/physiology , Female
6.
Curr Biol ; 34(16): 3820-3829.e5, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39079532

ABSTRACT

Gametes are produced via meiosis, a specialized cell division associated with frequent errors that cause birth defects and infertility. Uniquely in meiosis I, homologous chromosomes segregate to opposite poles, usually requiring their linkage by chiasmata, the products of crossover recombination.1 The spindle checkpoint delays cell-cycle progression until all chromosomes are properly attached to microtubules,2 but the steps leading to the capture and alignment of chromosomes on the meiosis I spindle remain poorly understood. In budding yeast meiosis I, Mad2 and Mad3BUBR1 are equally important for spindle checkpoint delay, but biorientation of homologs on the meiosis I spindle requires Mad2, but not Mad3BUBR1.3,4 Here we reveal the distinct functions of Mad2 and Mad3BUBR1 in meiosis I chromosome segregation. Mad2 promotes the prophase to metaphase I transition, while Mad3BUBR1 associates with the TOGL1 domain of Stu1CLASP, a conserved plus-end microtubule protein that is important for chromosome capture onto the spindle. Homologous chromosome pairs that are proficient in crossover formation but fail to biorient rely on Mad3BUBR1-Stu1CLASP to ensure their efficient attachment to microtubules and segregation during meiosis I. Furthermore, we show that Mad3BUBR1-Stu1CLASP are essential to rescue the segregation of mini-chromosomes lacking crossovers. Our findings define a new pathway ensuring microtubule-dependent chromosome capture and demonstrate that spindle checkpoint proteins safeguard the fidelity of chromosome segregation both by actively promoting chromosome alignment and by delaying cell-cycle progression until this has occurred.


Subject(s)
Cell Cycle Proteins , Chromosome Segregation , M Phase Cell Cycle Checkpoints , Meiosis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Spindle Apparatus , Meiosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Chromosome Segregation/physiology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , M Phase Cell Cycle Checkpoints/physiology , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Mad2 Proteins/metabolism , Mad2 Proteins/genetics , Microtubules/metabolism
7.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960623

ABSTRACT

In many animal species, the oocyte meiotic spindle, which is required for chromosome segregation, forms without centrosomes. In some systems, Ran-GEF on chromatin initiates spindle assembly. We found that in Caenorhabditis elegans oocytes, endogenously-tagged Ran-GEF dissociates from chromatin during spindle assembly but re-associates during meiotic anaphase. Meiotic spindle assembly occurred after auxin-induced degradation of Ran-GEF, but anaphase I was faster than controls and extrusion of the first polar body frequently failed. In search of a possible alternative pathway for spindle assembly, we found that soluble tubulin concentrates in the nuclear volume during germinal vesicle breakdown. We found that the concentration of soluble tubulin in the metaphase spindle region is enclosed by ER sheets which exclude cytoplasmic organelles including mitochondria and yolk granules. Measurement of the volume occupied by yolk granules and mitochondria indicated that volume exclusion would be sufficient to explain the concentration of tubulin in the spindle volume. We suggest that this concentration of soluble tubulin may be a redundant mechanism promoting spindle assembly near chromosomes.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Meiosis , Oocytes , Tubulin , ran GTP-Binding Protein , Animals , Anaphase , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Chromatin/metabolism , Chromosome Segregation , Guanosine Triphosphate/metabolism , Meiosis/physiology , Oocytes/metabolism , Prometaphase , ran GTP-Binding Protein/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
8.
Theriogenology ; 226: 335-342, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38959844

ABSTRACT

Extracellular signal-regulated protein kinase 5 (Erk5), a member of the mitogen-activated protein kinase (MAPK) family, is ubiquitously expressed in all eukaryotic cells and is implicated in the various mitotic processes such as cell survival, proliferation, migration, and differentiation. However, the potential functional roles of Erk5 in oocyte meiosis have not been fully determined. In this study, we document that ERK5 participates in the meiotic maturation of mouse oocytes by regulating the spindle assembly to ensure the meiotic progression. We unexpectedly found that phosphorylated ERK5 was localized in the spindle pole region at metaphase I and II stages by immunostaining analysis. Inhibition of ERK5 activity using its specific inhibitor XMD8-92 dramatically reduced the incidence of first polar body extrusion. In addition, inhibition of ERK5 evoked the spindle assembly checkpoint to arrest oocytes at metaphase I stage by impairing the spindle assembly, chromosome alignment and kinetochore-microtubule attachment. Mechanically, over-strengthened microtubule stability was shown to disrupt the microtubule dynamics and thus compromise the spindle assembly in ERK5-inhibited oocytes. Conversely, overexpression of ERK5 caused decreased level of acetylated α-tubulin and spindle defects. Collectively, we conclude that ERK5 plays an important role in the oocyte meiotic maturation by regulating microtubule dynamics and spindle assembly.


Subject(s)
Meiosis , Mitogen-Activated Protein Kinase 7 , Oocytes , Spindle Apparatus , Animals , Oocytes/physiology , Meiosis/physiology , Mice , Spindle Apparatus/physiology , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Female
9.
Front Endocrinol (Lausanne) ; 15: 1365260, 2024.
Article in English | MEDLINE | ID: mdl-38887270

ABSTRACT

Anti-Müllerian hormone (AMH) is a key paracrine/autocrine factor regulating folliculogenesis in the postnatal ovary. As antral follicles mature to the preovulatory stage, AMH production tends to be limited to cumulus cells. Therefore, the present study investigated the role of cumulus cell-derived AMH in supporting maturation and competence of the enclosed oocyte. Cumulus-oocyte complexes (COCs) were isolated from antral follicles of rhesus macaque ovaries for in vitro maturation with or without AMH depletion. Oocyte meiotic status and embryo cleavage after in vitro fertilization were assessed. In vitro maturation with AMH depletion was also performed using COCs from antral follicles of human ovarian tissue. Oocyte maturation and morphology were evaluated. The direct AMH action on mural granulosa cells of the preovulatory follicle was further assessed using human granulosa cells cultured with or without AMH supplementation. More macaque COCs produced metaphase II oocytes with AMH depletion than those of the control culture. However, preimplantation embryonic development after in vitro fertilization was comparable between oocytes derived from COCs cultured with AMH depletion and controls. Oocytes resumed meiosis in human COCs cultured with AMH depletion and exhibited a typical spindle structure. The confluency and cell number decreased in granulosa cells cultured with AMH supplementation relative to the control culture. AMH treatment did not induce cell death in cultured human granulosa cells. Data suggest that reduced AMH action in COCs could be beneficial for oocyte maturation. Cumulus cell-derived AMH is not essential for supporting oocyte competence or mural granulosa cell viability.


Subject(s)
Anti-Mullerian Hormone , Cumulus Cells , In Vitro Oocyte Maturation Techniques , Macaca mulatta , Oocytes , Anti-Mullerian Hormone/metabolism , Oocytes/metabolism , Oocytes/cytology , Oocytes/drug effects , Female , Cumulus Cells/metabolism , Cumulus Cells/cytology , Cumulus Cells/drug effects , Animals , Humans , In Vitro Oocyte Maturation Techniques/methods , Oogenesis/physiology , Oogenesis/drug effects , Cells, Cultured , Fertilization in Vitro/methods , Meiosis/physiology , Meiosis/drug effects , Granulosa Cells/metabolism , Granulosa Cells/cytology , Ovarian Follicle/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Embryonic Development/physiology
10.
Mol Biol Cell ; 35(8): ar105, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38865189

ABSTRACT

The reductional division of meiosis I requires the separation of chromosome pairs towards opposite poles. We have previously implicated the outer kinetochore protein SPC105R/KNL1 in driving meiosis I chromosome segregation through lateral attachments to microtubules and coorientation of sister centromeres. To identify the domains of SPC105R that are critical for meiotic chromosome segregation, an RNAi-resistant gene expression system was developed. We found that the SPC105R C-terminal domain (aa 1284-1960) is necessary and sufficient for recruiting NDC80 to the kinetochore and building the outer kinetochore. Furthermore, the C-terminal domain recruits BUBR1, which in turn recruits the cohesion protection proteins MEI-S332 and PP2A. Of the remaining 1283 amino acids, we found the first 473 are most important for meiosis. The first 123 amino acids of the N-terminal half of SPC105R contain the conserved SLRK and RISF motifs that are targets of PP1 and Aurora B kinase and are most important for regulating the stability of microtubule attachments and maintaining metaphase I arrest. The region between amino acids 124 and 473 are required for lateral microtubule attachments and biorientation of homologues, which are critical for accurate chromosome segregation in meiosis I.


Subject(s)
Chromosome Segregation , Drosophila Proteins , Kinetochores , Meiosis , Microtubules , Oocytes , Kinetochores/metabolism , Animals , Meiosis/physiology , Oocytes/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Female , Centromere/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Aurora Kinase B/metabolism , Aurora Kinase B/genetics
11.
Sheng Li Xue Bao ; 76(3): 438-446, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38939938

ABSTRACT

Oocyte maturation and early embryonic development are key steps in the reproductive physiology of female mammals, and any error in this process can adversely affect reproductive development. Recent studies have shown that epigenetic modifications of histones play important roles in the regulation of oocyte meiosis and quality assurance of early embryonic development. Histone deacetylase 11 (HDAC11) is the smallest known member of the histone deacetylases (HDACs) family, and inhibition of HDAC11 activity significantly suppresses the rate of oocyte maturation, as well as the development of 8-cell and blastocyst embryos at the embryonic stage. This paper focuses on recent progress on the important role of HDAC11 in the regulation of mammalian oocyte maturation and early embryonic development, hoping to gain insights into the key roles played by epitope-modifying proteins represented by HDAC11 in the regulation of mammalian reproduction and their molecular mechanisms.


Subject(s)
Embryonic Development , Histone Deacetylases , Oocytes , Animals , Oocytes/physiology , Embryonic Development/physiology , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Histone Deacetylases/genetics , Female , Humans , Oogenesis/physiology , Mammals/embryology , Meiosis/physiology
12.
Mol Reprod Dev ; 91(6): e23763, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38895803

ABSTRACT

Estrogen is an important hormone that plays a role in regulating follicle development and oocyte maturation. Transzonal projections (TZPs) act as communication bridges between follicle somatic cells and oocytes, and their dynamic changes are critical for oocyte development and maturation. However, the roles and mechanisms of estrogen in regulating TZPs during follicular development are not yet understood. We found that the proportion of oocytes spontaneously resuming meiosis increases as the follicle grows, which is accompanied by rising estrogen levels in follicles and decreasing TZPs in cumulus-oocyte complex. To further explore the effect of elevated estrogen levels on TZP assembly, additional estrogen was added to the culture system. The increased estrogen level significantly decreased the mRNA and protein expression levels of TZP assembly-related genes. Subsequent research revealed that TZP regulation by estrogen was mediated by the membrane receptor GPER and downstream ERK1/2 signaling pathway. In summary, our study suggests that estrogen may regulate goat oocyte meiosis arrest by decreasing TZP numbers via estrogen-mediated GPER activation during follicle development.


Subject(s)
Cumulus Cells , Estrogens , Goats , Oocytes , Ovarian Follicle , Receptors, Estrogen , Receptors, G-Protein-Coupled , Animals , Oocytes/metabolism , Oocytes/cytology , Female , Cumulus Cells/metabolism , Cumulus Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Estrogen/metabolism , Estrogens/metabolism , Ovarian Follicle/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/cytology , Meiosis/physiology , MAP Kinase Signaling System/physiology
13.
Biol Res ; 57(1): 36, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822414

ABSTRACT

BACKGROUND: Helicase for meiosis 1 (HFM1), a putative DNA helicase expressed in germ-line cells, has been reported to be closely associated with premature ovarian insufficiency (POI). However, the underlying molecular mechanism has not been clearly elucidated. The aim of this study was to investigate the function of HFM1 in the first meiotic prophase of mouse oocytes. RESULTS: The results suggested that the deficiency of HFM1 resulting in increased apoptosis and depletion of oocytes in mice, while the oocytes were arrested in the pachytene stage of the first meiotic prophase. In addition, impaired DNA double-strand break repair and disrupted synapsis were observed in the absence of HFM1. Further investigation revealed that knockout of HFM1 promoted ubiquitination and degradation of FUS protein mediated by FBXW11. Additionally, the depletion of HFM1 altered the intranuclear localization of FUS and regulated meiotic- and oocyte development-related genes in oocytes by modulating the expression of BRCA1. CONCLUSIONS: These findings elaborated that the critical role of HFM1 in orchestrating the regulation of DNA double-strand break repair and synapsis to ensure meiosis procession and primordial follicle formation. This study provided insights into the pathogenesis of POI and highlighted the importance of HFM1 in maintaining proper meiotic function in mouse oocytes.


Subject(s)
Meiotic Prophase I , Oocytes , Ubiquitination , Animals , Female , Mice , Apoptosis/physiology , DNA Breaks, Double-Stranded , DNA Repair/physiology , Meiosis/physiology , Meiotic Prophase I/physiology , Mice, Knockout , Oocytes/metabolism , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics
14.
FASEB J ; 38(13): e23750, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38888878

ABSTRACT

Kif16A, a member of the kinesin-3 family of motor proteins, has been shown to play crucial roles in inducing mitotic arrest, apoptosis, and mitotic cell death. However, its roles during oocyte meiotic maturation have not been fully defined. In this study, we report that Kif16A exhibits unique accumulation on the spindle apparatus and colocalizes with microtubule fibers during mouse oocyte meiotic maturation. Targeted depletion of Kif16A using gene-targeting siRNA disrupts the progression of the meiotic cell cycle. Furthermore, Kif16A depletion leads to aberrant spindle assembly and chromosome misalignment in oocytes. Our findings also indicate that Kif16A depletion reduces tubulin acetylation levels and compromises microtubule resistance to depolymerizing drugs, suggesting its crucial role in microtubule stability maintenance. Notably, we find that the depletion of Kif16A results in a notably elevated incidence of defective kinetochore-microtubule attachments and the absence of BubR1 localization at kinetochores, suggesting a critical role for Kif16A in the activation of the spindle assembly checkpoint (SAC) activity. Additionally, we observe that Kif16A is indispensable for proper actin filament distribution, thereby impacting spindle migration. In summary, our findings demonstrate that Kif16A plays a pivotal role in regulating microtubule and actin dynamics crucial for ensuring both spindle assembly and migration during mouse oocyte meiotic maturation.


Subject(s)
Kinesins , Meiosis , Microtubules , Oocytes , Spindle Apparatus , Animals , Kinesins/metabolism , Kinesins/genetics , Meiosis/physiology , Oocytes/metabolism , Microtubules/metabolism , Mice , Spindle Apparatus/metabolism , Female , Actins/metabolism , Kinetochores/metabolism
15.
Mol Hum Reprod ; 30(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38870523

ABSTRACT

Advanced maternal age is associated with a decline in oocyte quality, which often leads to reproductive failure in humans. However, the mechanisms behind this age-related decline remain unclear. To gain insights into this phenomenon, we applied plexDIA, a multiplexed data-independent acquisition, single-cell mass spectrometry method, to analyze the proteome of oocytes from both young women and women of advanced maternal age. Our findings primarily revealed distinct proteomic profiles between immature fully grown germinal vesicle and mature metaphase II oocytes. Importantly, we further show that a woman's age is associated with changes in her oocyte proteome. Specifically, when compared to oocytes obtained from young women, advanced maternal age oocytes exhibited lower levels of the proteasome and TRiC complex, as well as other key regulators of proteostasis and meiosis. This suggests that aging adversely affects the proteostasis and meiosis networks in human oocytes. The proteins identified in this study hold potential as targets for improving oocyte quality and may guide future studies into the molecular processes underlying oocyte aging.


Subject(s)
Maternal Age , Meiosis , Oocytes , Proteome , Proteomics , Proteostasis , Single-Cell Analysis , Humans , Oocytes/metabolism , Oocytes/cytology , Female , Meiosis/physiology , Adult , Proteomics/methods , Single-Cell Analysis/methods , Proteome/metabolism , Proteasome Endopeptidase Complex/metabolism , Middle Aged
16.
FASEB J ; 38(13): e23739, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38884157

ABSTRACT

Arf6 is a member of ADP-ribosylation factor (Arf) family, which is widely implicated in the regulation of multiple physiological processes including endocytic recycling, cytoskeletal organization, and membrane trafficking during mitosis. In this study, we investigated the potential relationship between Arf6 and aging-related oocyte quality, and its roles on organelle rearrangement and cytoskeleton dynamics in porcine oocytes. Arf6 expressed in porcine oocytes throughout meiotic maturation, and it decreased in aged oocytes. Disruption of Arf6 led to the failure of cumulus expansion and polar body extrusion. Further analysis indicated that Arf6 modulated ac-tubulin for meiotic spindle organization and microtubule stability. Besides, Arf6 regulated cofilin phosphorylation and fascin for actin assembly, which further affected spindle migration, indicating the roles of Arf6 on cytoskeleton dynamics. Moreover, the lack of Arf6 activity caused the dysfunction of Golgi and ER for protein synthesis and signal transduction. Mitochondrial dysfunction was also observed in Arf6-deficient porcine oocytes, which was supported by the increased ROS level and abnormal membrane potential. In conclusion, our results reported that insufficient Arf6 was related to aging-induced oocyte quality decline through spindle organization, actin assembly, and organelle rearrangement in porcine oocytes.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Oocytes , Animals , Oocytes/metabolism , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Swine , Female , Meiosis/physiology , Spindle Apparatus/metabolism , Aging/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
17.
Proc Natl Acad Sci U S A ; 121(27): e2317316121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917013

ABSTRACT

A dispersed cytoplasmic distribution of mitochondria is a hallmark of normal cellular organization. Here, we have utilized the expression of exogenous Trak2 in mouse oocytes and embryos to disrupt the dispersed distribution of mitochondria by driving them into a large cytoplasmic aggregate. Our findings reveal that aggregated mitochondria have minimal impact on asymmetric meiotic cell divisions of the oocyte. In contrast, aggregated mitochondria during the first mitotic division result in daughter cells with unequal sizes and increased micronuclei. Further, in two-cell embryos, microtubule-mediated centering properties of the mitochondrial aggregate prevent nuclear centration, distort nuclear shape, and inhibit DNA synthesis and the onset of embryonic transcription. These findings demonstrate the motor protein-mediated distribution of mitochondria throughout the cytoplasm is highly regulated and is an essential feature of cytoplasmic organization to ensure optimal cell function.


Subject(s)
Blastocyst , Cell Nucleus , Mitochondria , Oocytes , Animals , Mitochondria/metabolism , Blastocyst/metabolism , Blastocyst/cytology , Mice , Cell Nucleus/metabolism , Oocytes/metabolism , Oocytes/cytology , Female , Embryonic Development/physiology , Microtubules/metabolism , Mitosis , Meiosis/physiology
18.
Theriogenology ; 225: 43-54, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38788628

ABSTRACT

Extensive research has been conducted on the role of CXCR3 in immune responses and inflammation. However, the role of CXCR3 in the reproductive system, particularly in oocyte development, remains unknown. In this study, we present findings on the involvement of CXCR3 in the meiotic division process of mouse oocytes. We found CXCR3 was expressed consistently throughout the entire maturation process of mouse oocyte. Inhibition of CXCR3 impaired the asymmetric division of oocyte, while the injection of Cxcr3 mRNA was capable of restoring these defects. Further study showed that inhibition of CXCR3 perturbed spindle migration by affecting LIMK/cofilin pathway-mediated actin remodeling. Knockout of CXCR3 led to an upregulation of actin-binding protein and an increased ATP level in GV-stage oocytes, while maintaining normal actin dynamics during the process of meiosis. Additionally, we noticed the expression level of DYNLT1 is markedly elevated in CXCR3-null oocytes. DYNLT1 bound with the Arp2/3 complex, and knockdown of DYNLT1 in CXCR3-null oocytes impaired the organization of cytoplasmic actin, suggesting the regulatory role of DYNLT1 in actin organization, and the compensatory expression of DYNLT1 may contribute to maintain normal actin dynamics in CXCR3-knockout oocytes. In summary, our findings provide insights into the intricate network of actin dynamics associated with CXCR3 during oocyte meiosis.


Subject(s)
Actins , Oocytes , Receptors, CXCR3 , Animals , Oocytes/metabolism , Oocytes/physiology , Mice , Actins/metabolism , Actins/genetics , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Female , Meiosis/physiology , Mice, Knockout
19.
Zool Res ; 45(3): 601-616, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38766744

ABSTRACT

Meiosis is a highly complex process significantly influenced by transcriptional regulation. However, studies on the mechanisms that govern transcriptomic changes during meiosis, especially in prophase I, are limited. Here, we performed single-cell ATAC-seq of human testis tissues and observed reprogramming during the transition from zygotene to pachytene spermatocytes. This event, conserved in mice, involved the deactivation of genes associated with meiosis after reprogramming and the activation of those related to spermatogenesis before their functional onset. Furthermore, we identified 282 transcriptional regulators (TRs) that underwent activation or deactivation subsequent to this process. Evidence suggested that physical contact signals from Sertoli cells may regulate these TRs in spermatocytes, while secreted ENHO signals may alter metabolic patterns in these cells. Our results further indicated that defective transcriptional reprogramming may be associated with non-obstructive azoospermia (NOA). This study revealed the importance of both physical contact and secreted signals between Sertoli cells and germ cells in meiotic progression.


Subject(s)
Cell Communication , Meiosis , Animals , Male , Mice , Meiosis/physiology , Humans , Sertoli Cells/metabolism , Sertoli Cells/physiology , Testis/metabolism , Testis/cytology , Spermatogenesis/physiology , Gene Expression Regulation , Azoospermia/genetics , Transcription, Genetic , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , Single-Cell Gene Expression Analysis
20.
Reprod Biomed Online ; 49(1): 103976, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733676

ABSTRACT

RESEARCH QUESTION: Can immature oocytes vitrified and warmed using a short protocol survive and resume meiosis? DESIGN: This study examined modifications of oocyte vitrification and warming protocols that reduce the length of exposure to vitrification and warming solutions. In total, 561 germinal vesicles and 218 metaphase I oocytes that were immature at oocyte retrieval were vitrified at room temperature for 2 min. Warming was performed at 37°C for 2 min. Resumption of meiotic activity was evaluated after 24 and 48 h of culture. Two different commercially available vitrification and warming kits were used for comparison. RESULTS: Ninety-five percent of germinal vesicles survived, with no difference observed between the kits. The survival of metaphase I oocytes was, on average, 95.4% and did not differ significantly between the kits. Of the 533 germinal vesicles that survived, 491 converted to metaphase I oocytes (92.1%). After culture for 48 h, 54.4% converted to metaphase II oocytes. In addition, of the 208 metaphase I oocytes that survived warming, 84.1% converted to metaphase II oocytes after 24 h of culture. These maturation rates were similar to those of non-vitrified oocytes. CONCLUSIONS: Vitrification and warming of oocytes at different nuclear maturation stages can be performed with 2 min of exposure to hypertonic solution and 2 min of exposure to hypotonic solution, respectively. This approach reduces exposure of the oocytes to room temperature during dehydration and rehydration. Warming in 0.5M sucrose helps to maintain and support the potential of oocytes to resume nuclear meiotic activity, and conversion from germinal vesicles to metaphase I and metaphase II oocytes.


Subject(s)
Cryopreservation , Meiosis , Oocytes , Vitrification , Oocytes/cytology , Oocytes/physiology , Humans , Meiosis/physiology , Female , Cryopreservation/methods , Cell Survival , In Vitro Oocyte Maturation Techniques/methods , Adult
SELECTION OF CITATIONS
SEARCH DETAIL