Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.179
Filter
1.
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
2.
Nat Cell Biol ; 26(6): 962-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839978

ABSTRACT

Dynamic epigenomic reprogramming occurs during mammalian oocyte maturation and early development. However, the underlying transcription circuitry remains poorly characterized. By mapping cis-regulatory elements using H3K27ac, we identified putative enhancers in mouse oocytes and early embryos distinct from those in adult tissues, enabling global transitions of regulatory landscapes around fertilization and implantation. Gene deserts harbour prevalent putative enhancers in fully grown oocytes linked to oocyte-specific genes and repeat activation. Embryo-specific enhancers are primed before zygotic genome activation and are restricted by oocyte-inherited H3K27me3. Putative enhancers in oocytes often manifest H3K4me3, bidirectional transcription, Pol II binding and can drive transcription in STARR-seq and a reporter assay. Finally, motif analysis of these elements identified crucial regulators of oogenesis, TCF3 and TCF12, the deficiency of which impairs activation of key oocyte genes and folliculogenesis. These data reveal distinctive regulatory landscapes and their interacting transcription factors that underpin the development of mammalian oocytes and early embryos.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Oocytes , Oogenesis , Animals , Oocytes/metabolism , Female , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Oogenesis/genetics , Mice , Histones/metabolism , Histones/genetics , Embryo, Mammalian/metabolism , Mice, Inbred C57BL , Embryonic Development/genetics , Ovarian Follicle/metabolism , Mice, Knockout
3.
C R Biol ; 347: 45-52, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888193

ABSTRACT

Fertility is declining worldwide and many couples are turning towards assisted reproductive technologies (ART) to conceive babies. Organisms that propagate via sexual reproduction often come from the fusion between two gametes, an oocyte and a sperm, whose qualities seem to be decreasing in the human species. Interestingly, while the sperm mostly transmits its haploid genome, the oocyte transmits not only its haploid set of chromosomes but also its huge cytoplasm to its progeny. This is what can be defined as the maternal inheritance composed of chromosomes, organelles, lipids, metabolites, proteins and RNAs. To decipher the decline in oocyte quality, it is essential to explore the nature of the maternal inheritance, and therefore study the last stages of murine oogenesis, namely the end of oocyte growth followed by the two meiotic divisions. These divisions are extremely asymmetric in terms of the size of the daughter cells, allowing to preserve the maternal inheritance accumulated during oocyte growth within these huge cells to support early embryo development. Studies performed in Marie-Hélène Verlhac's lab have allowed to discover the unprecedented impact of original acto-myosin based mechanisms in the constitution as well as the preservation of this maternal inheritance and the consequences when these processes go awry.


La fécondité diminue mondialement et de nombreux couples se tournent vers les techniques de procréation médicalement assistée (PMA) pour concevoir des bébés. Les organismes se propageant par reproduction sexuée sont souvent issus de la fusion de deux gamètes, un ovocyte et un spermatozoïde, dont les qualités semblent diminuer dans l'espèce humaine. Si le spermatozoïde transmet principalement son génome haploïde, l'ovocyte transmet à sa progéniture non seulement son lot haploïde de chromosomes, mais aussi son immense cytoplasme. C'est ce que l'on peut définir comme l'héritage maternel, composé de chromosomes, d'organelles, de lipides, de métabolites, de protéines et d'ARNs. Pour comprendre la baisse de qualité des ovocytes, il est essentiel d'explorer la nature de cet héritage maternel, et donc d'étudier les dernières étapes de l'ovogenèse murine, à savoir la fin de la croissance ovocytaire suivie des deux divisions méiotiques. Ces divisions sont extrêmement asymétriques par la taille des cellules filles engendrées, ce qui permet de préserver l'héritage maternel accumulé pendant la croissance de cette énorme cellule, l'ovocyte, pour soutenir le développement précoce de l'embryon. Les études menées dans le laboratoire de Marie-Hélène Verlhac ont permis de découvrir l'impact sans précédent de mécanismes originaux dépendant de l'acto-myosine dans la constitution et la préservation de cet héritage maternel, ainsi que les conséquences des erreurs dans ces processus.


Subject(s)
Maternal Inheritance , Oocytes , Animals , Female , Humans , Mice , Maternal Inheritance/genetics , Meiosis , Oogenesis/genetics
4.
Nat Commun ; 15(1): 5331, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909026

ABSTRACT

Cytoplasmic polyadenylation plays a vital role in gametogenesis; however, the participating enzymes and substrates in mammals remain unclear. Using knockout and knock-in mouse models, we describe the essential role of four TENT5 poly(A) polymerases in mouse fertility and gametogenesis. TENT5B and TENT5C play crucial yet redundant roles in oogenesis, with the double knockout of both genes leading to oocyte degeneration. Additionally, TENT5B-GFP knock-in females display a gain-of-function infertility effect, with multiple chromosomal aberrations in ovulated oocytes. TENT5C and TENT5D both regulate different stages of spermatogenesis, as shown by the sterility in males following the knockout of either gene. Finally, Tent5a knockout substantially lowers fertility, although the underlying mechanism is not directly related to gametogenesis. Through direct RNA sequencing, we discovered that TENT5s polyadenylate mRNAs encoding endoplasmic reticulum-targeted proteins essential for gametogenesis. Sequence motif analysis and reporter mRNA assays reveal that the presence of an endoplasmic reticulum-leader sequence represents the primary determinant of TENT5-mediated regulation.


Subject(s)
Gametogenesis , Mice, Knockout , Polyadenylation , RNA, Messenger , Spermatogenesis , Animals , Female , Male , RNA, Messenger/metabolism , RNA, Messenger/genetics , Mice , Spermatogenesis/genetics , Gametogenesis/genetics , Oogenesis/genetics , Polynucleotide Adenylyltransferase/metabolism , Polynucleotide Adenylyltransferase/genetics , Oocytes/metabolism , Fertility/genetics , Mice, Inbred C57BL
5.
Genes Dev ; 38(9-10): 436-454, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38866556

ABSTRACT

Genome organization can regulate gene expression and promote cell fate transitions. The differentiation of germline stem cells (GSCs) to oocytes in Drosophila involves changes in genome organization mediated by heterochromatin and the nuclear pore complex (NPC). Heterochromatin represses germ cell genes during differentiation, and NPCs anchor these silenced genes to the nuclear periphery, maintaining silencing to allow for oocyte development. Surprisingly, we found that genome organization also contributes to NPC formation, mediated by the transcription factor Stonewall (Stwl). As GSCs differentiate, Stwl accumulates at boundaries between silenced and active gene compartments. Stwl at these boundaries plays a pivotal role in transitioning germ cell genes into a silenced state and activating a group of oocyte genes and nucleoporins (Nups). The upregulation of these Nups during differentiation is crucial for NPC formation and further genome organization. Thus, cross-talk between genome architecture and NPCs is essential for successful cell fate transitions.


Subject(s)
Cell Differentiation , Drosophila Proteins , Genome, Insect , Nuclear Pore , Oogenesis , Animals , Oogenesis/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Cell Differentiation/genetics , Nuclear Pore/metabolism , Nuclear Pore/genetics , Genome, Insect/genetics , Gene Expression Regulation, Developmental/genetics , Female , Drosophila melanogaster/genetics , Oocytes/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore Complex Proteins/genetics
6.
Article in English | MEDLINE | ID: mdl-38862425

ABSTRACT

Gametogenesis plays an important role in the reproduction and evolution of species. The transcriptomic and epigenetic alterations in this process can influence the reproductive capacity, fertilization, and embryonic development. The rapidly increasing single-cell studies have provided valuable multi-omics resources. However, data from different layers and sequencing platforms have not been uniformed and integrated, which greatly limits their use for exploring the molecular mechanisms that underlie oogenesis and spermatogenesis. Here, we develop GametesOmics, a comprehensive database that integrates the data of gene expression, DNA methylation, and chromatin accessibility during oogenesis and spermatogenesis in humans and mice. GametesOmics provides a user-friendly website and various tools, including Search and Advanced Search for querying the expression and epigenetic modification(s) of each gene; Tools with Differentially expressed gene (DEG) analysis for identifying DEGs, Correlation analysis for demonstrating the genetic and epigenetic changes, Visualization for displaying single-cell clusters and screening marker genes as well as master transcription factors (TFs), and MethylView for studying the genomic distribution of epigenetic modifications. GametesOmics also provides Genome Browser and Ortholog for tracking and comparing gene expression, DNA methylation, and chromatin accessibility between humans and mice. GametesOmics offers a comprehensive resource for biologists and clinicians to decipher the cell fate transition in germ cell development, and can be accessed at http://gametesomics.cn/.


Subject(s)
DNA Methylation , Databases, Genetic , Gametogenesis , Animals , Humans , Mice , Gametogenesis/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Male , Germ Cells/metabolism , Female , Spermatogenesis/genetics , Oogenesis/genetics , Genomics/methods , Multiomics
7.
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
8.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891872

ABSTRACT

Species of the genus Drosophila have served as favorite models in speciation studies; however, genetic factors of interspecific reproductive incompatibility are under-investigated. Here, we performed an analysis of hybrid female sterility by crossing Drosophila melanogaster females and Drosophila simulans males. Using transcriptomic data analysis and molecular, cellular, and genetic approaches, we analyzed differential gene expression, transposable element (TE) activity, piRNA biogenesis, and functional defects of oogenesis in hybrids. Premature germline stem cell loss was the most prominent defect of oogenesis in hybrid ovaries. Because of the differential expression of genes encoding piRNA pathway components, rhino and deadlock, the functional RDCmel complex in hybrid ovaries was not assembled. However, the activity of the RDCsim complex was maintained in hybrids independent of the genomic origin of piRNA clusters. Despite the identification of a cohort of overexpressed TEs in hybrid ovaries, we found no evidence that their activity can be considered the main cause of hybrid sterility. We revealed a complicated pattern of Vasa protein expression in the hybrid germline, including partial AT-chX piRNA targeting of the vasasim allele and a significant zygotic delay in vasamel expression. We arrived at the conclusion that the hybrid sterility phenotype was caused by intricate multi-locus differences between the species.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Drosophila simulans , RNA, Small Interfering , Animals , Female , Drosophila melanogaster/genetics , Male , Drosophila simulans/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , RNA, Small Interfering/genetics , DNA Transposable Elements/genetics , Ovary/metabolism , Hybridization, Genetic , Oogenesis/genetics , Infertility/genetics , Crosses, Genetic , DEAD-box RNA Helicases
9.
Nat Commun ; 15(1): 5248, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898112

ABSTRACT

Reproductive success relies on proper establishment and maintenance of biological sex. In many animals, including mammals, the primary gonad is initially ovary biased. We previously showed the RNA binding protein (RNAbp), Rbpms2, is required for ovary fate in zebrafish. Here, we identified Rbpms2 targets in oocytes (Rbpms2-bound oocyte RNAs; rboRNAs). We identify Rbpms2 as a translational regulator of rboRNAs, which include testis factors and ribosome biogenesis factors. Further, genetic analyses indicate that Rbpms2 promotes nucleolar amplification via the mTorc1 signaling pathway, specifically through the mTorc1-activating Gap activity towards Rags 2 (Gator2) component, Missing oocyte (Mios). Cumulatively, our findings indicate that early gonocytes are in a dual poised, bipotential state in which Rbpms2 acts as a binary fate-switch. Specifically, Rbpms2 represses testis factors and promotes oocyte factors to promote oocyte progression through an essential Gator2-mediated checkpoint, thereby integrating regulation of sexual differentiation factors and nutritional availability pathways in zebrafish oogenesis.


Subject(s)
Oocytes , Oogenesis , RNA-Binding Proteins , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Female , Oocytes/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Oogenesis/genetics , Male , Ovary/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Signal Transduction , Gene Expression Regulation, Developmental , Testis/metabolism , Nutrients/metabolism
10.
Mol Biol Evol ; 41(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38880992

ABSTRACT

Although evolution is driven by changes in how regulatory pathways control development, we know little about the molecular details underlying these transitions. The TRA-2 domain that mediates contact with TRA-1 is conserved in Caenorhabditis. By comparing the interaction of these proteins in two species, we identified a striking change in how sexual development is controlled. Identical mutations in this domain promote oogenesis in Caenorhabditis elegans but promote spermatogenesis in Caenorhabditis briggsae. Furthermore, the effects of these mutations involve the male-promoting gene fem-3 in C. elegans but are independent of fem-3 in C. briggsae. Finally, reciprocal mutations in these genes show that C. briggsae TRA-2 binds TRA-1 to prevent expression of spermatogenesis regulators. By contrast, in C. elegans TRA-1 sequesters TRA-2 in the germ line, allowing FEM-3 to initiate spermatogenesis. Thus, we propose that the flow of information within the sex determination pathway has switched directions during evolution. This result has important implications for how evolutionary change can occur.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Sex Determination Processes , Spermatogenesis , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Male , Spermatogenesis/genetics , Female , Caenorhabditis/genetics , Biological Evolution , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mutation , Oogenesis/genetics , Evolution, Molecular , Self-Fertilization , DNA-Binding Proteins , Transcription Factors
11.
Int J Mol Sci ; 25(9)2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38732042

ABSTRACT

Numerous post-translational modifications are involved in oocyte maturation and embryo development. Recently, lactylation has emerged as a novel epigenetic modification implicated in the regulation of diverse cellular processes. However, it remains unclear whether lactylation occurs during oocyte maturation and embryo development processes. Herein, the lysine lactylation (Kla) modifications were determined during mouse oocyte maturation and early embryo development by immunofluorescence staining. Exogenous lactate was supplemented to explore the consequences of modulating histone lactylation levels on oocyte maturation and embryo development processes by transcriptomics. Results demonstrated that lactylated proteins are widely present in mice with tissue- and cell-specific distribution. During mouse oocyte maturation, immunofluorescence for H3K9la, H3K14la, H4K8la, and H4K12la was most intense at the germinal vesicle (GV) stage and subsequently weakened or disappeared. Further, supplementing the culture medium with 10 mM sodium lactate elevated both the oocyte maturation rate and the histone Kla levels in GV oocytes, and there were substantial increases in Kla levels in metaphase II (MII) oocytes. It altered the transcription of molecules involved in oxidative phosphorylation. Moreover, histone lactylation levels changed dynamically during mouse early embryogenesis. Sodium lactate at 10 mM enhanced early embryo development and significantly increased lactylation, while impacting glycolytic gene transcription. This study reveals the roles of lactylation during oocyte maturation and embryo development, providing new insights to improving oocyte maturation and embryo quality.


Subject(s)
Embryonic Development , Histones , Oocytes , Protein Processing, Post-Translational , Animals , Histones/metabolism , Oocytes/metabolism , Mice , Embryonic Development/genetics , Female , Oogenesis , Lysine/metabolism , In Vitro Oocyte Maturation Techniques , Gene Expression Regulation, Developmental
12.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38735763

ABSTRACT

Follicle-stimulating hormone (FSH) binds to its membrane receptor (FSHR) in granulosa cells to activate various signal transduction pathways and drive the gonadotropin-dependent phase of folliculogenesis. Both FSH insufficiency (due to genetic or nongenetic factors) and FSH excess (as encountered with ovarian stimulation in assisted reproductive technology [ART]) can cause poor female reproductive outcomes, but the underlying molecular mechanisms remain elusive. Herein, we conducted single-follicle and single-oocyte RNA sequencing analysis along with other approaches in an ex vivo mouse folliculogenesis and oogenesis system to investigate the effects of different concentrations of FSH on key follicular events. Our study revealed that a minimum FSH threshold is required for follicle maturation into the high estradiol-secreting preovulatory stage, and such threshold is moderately variable among individual follicles between 5 and 10 mIU/mL. FSH at 5, 10, 20, and 30 mIU/mL induced distinct expression patterns of follicle maturation-related genes, follicular transcriptomics, and follicular cAMP levels. RNA sequencing analysis identified FSH-stimulated activation of G proteins and downstream canonical and novel signaling pathways that may critically regulate follicle maturation, including the cAMP/PKA/CREB, PI3K/AKT/FOXO1, and glycolysis pathways. High FSH at 20 and 30 mIU/mL resulted in noncanonical FSH responses, including premature luteinization, high production of androgen and proinflammatory factors, and reduced expression of energy metabolism-related genes in oocytes. Together, this study improves our understanding of gonadotropin-dependent folliculogenesis and provides crucial insights into how high doses of FSH used in ART may impact follicular health, oocyte quality, pregnancy outcome, and systemic health.


Subject(s)
Follicle Stimulating Hormone , Ovarian Follicle , Transcriptome , Animals , Female , Follicle Stimulating Hormone/pharmacology , Mice , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Transcriptome/drug effects , Dose-Response Relationship, Drug , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Oogenesis/genetics , Signal Transduction/drug effects , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Cyclic AMP/metabolism
13.
Mol Hum Reprod ; 30(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38745364

ABSTRACT

The role of cumulus cells (CCs) in the acquisition of oocyte developmental competence is not yet fully understood. In a previous study, we matured cumulus-denuded fully-grown mouse oocytes to metaphase II (MII) on a feeder layer of CCs (FL-CCs) isolated from developmentally competent (FL-SN-CCs) or incompetent (FL-NSN-CCs) SN (surrounded nucleolus) or NSN (not surrounding nucleolus) oocytes, respectively. We observed that oocytes cultured on the former could develop into blastocysts, while those matured on the latter arrested at the 2-cell stage. To investigate the CC factors contributing to oocyte developmental competence, here we focused on the CCs' release into the medium of extracellular vesicles (EVs) and on their miRNA content. We found that, during the 15-h transition to MII, both FL-SN-CCs and FL-NSN-CCs release EVs that can be detected, by confocal microscopy, inside the zona pellucida (ZP) or the ooplasm. The majority of EVs are <200 nm in size, which is compatible with their ability to cross the ZP. Next-generation sequencing of the miRNome of FL-SN-CC versus FL-NSN-CC EVs highlighted 74 differentially expressed miRNAs, with 43 up- and 31 down-regulated. Although most of these miRNAs do not have known roles in the ovary, in silico functional analysis showed that seven of these miRNAs regulate 71 target genes with specific roles in meiosis resumption (N = 24), follicle growth (N = 23), fertilization (N = 1), and the acquisition of oocyte developmental competence (N = 23). Overall, our results indicate CC EVs as emerging candidates of the CC-to-oocyte communication axis and uncover a group of miRNAs as potential regulatory factors.


Subject(s)
Cumulus Cells , Extracellular Vesicles , MicroRNAs , Oocytes , Animals , Cumulus Cells/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Oocytes/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Mice , Female , In Vitro Oocyte Maturation Techniques , Oogenesis/genetics , Zona Pellucida/metabolism
14.
Reprod Biol Endocrinol ; 22(1): 52, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711160

ABSTRACT

BACKGROUND: Elevated FSH often occurs in women of advanced maternal age (AMA, age ≥ 35) and in infertility patients undergoing controlled ovarian stimulation (COS). There is controversy on whether high endogenous FSH contributes to infertility and whether high exogenous FSH adversely impacts patient pregnancy rates. METHODS: The senescence-accelerated mouse-prone-8 (SAMP8) model of female reproductive aging was employed to assess the separate impacts of age and high FSH activity on the percentages (%) of viable and mature ovulated oocytes recovered after gonadotropin treatment. Young and midlife mice were treated with the FSH analog equine chorionic gonadotropin (eCG) to model both endogenous FSH elevation and exogenous FSH elevation. Previously we showed the activin inhibitor ActRIIB:Fc increases oocyte quality by preventing chromosome and spindle misalignments. Therefore, ActRIIB:Fc treatment was performed in an effort to increase % oocyte viability and % oocyte maturation. RESULTS: The high FSH activity of eCG is ootoxic to ovulatory oocytes, with greater decreases in % viable oocytes in midlife than young mice. High FSH activity of eCG potently inhibits oocyte maturation, decreasing the % of mature oocytes to similar degrees in young and midlife mice. ActRIIB:Fc treatment does not prevent eCG ootoxicity, but it restores most oocyte maturation impeded by eCG. CONCLUSIONS: FSH ootoxicity to ovulatory oocytes and FSH maturation inhibition pose a paradox given the well-known pro-growth and pro-maturation activities of FSH in the earlier stages of oocyte growth. We propose the FOOT Hypothesis ("FSH OoToxicity Hypothesis), that FSH ootoxicity to ovulatory oocytes comprises a new driver of infertility and low pregnancy success rates in DOR women attempting spontaneous pregnancy and in COS/IUI patients, especially AMA women. We speculate that endogenous FSH elevation also contributes to reduced fecundity in these DOR and COS/IUI patients. Restoration of oocyte maturation by ActRIB:Fc suggests that activin suppresses oocyte maturation in vivo. This contrasts with prior studies showing activin A promotes oocyte maturation in vitro. Improved oocyte maturation with agents that decrease endogenous activin activity with high specificity may have therapeutic benefit for COS/IVF patients, COS/IUI patients, and DOR patients attempting spontaneous pregnancies.


Subject(s)
Activin Receptors, Type II , Oocytes , Animals , Female , Oocytes/drug effects , Mice , Activin Receptors, Type II/metabolism , Ovulation/drug effects , Chorionic Gonadotropin/pharmacology , Follicle Stimulating Hormone/blood , Oogenesis/drug effects , Ovulation Induction/methods , Immunoglobulin Fc Fragments/pharmacology , Aging/drug effects , Aging/physiology , Pregnancy , Activins
15.
Biomolecules ; 14(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38785929

ABSTRACT

Suppressor of deltex (Su(dx)) is a Drosophila melanogaster member of the NEDD4 family of the HECT domain E3 ubiquitin ligases. Su(dx) acts as a regulator of Notch endocytic trafficking, promoting Notch lysosomal degradation and the down-regulation of both ligand-dependent and ligand-independent signalling, the latter involving trafficking through the endocytic pathway and activation of the endo/lysosomal membrane. Mutations of Su(dx) result in developmental phenotypes in the Drosophila wing that reflect increased Notch signalling, leading to gaps in the specification of the wing veins, and Su(dx) functions to provide the developmental robustness of Notch activity to environmental temperature shifts. The full developmental functions of Su(dx) are unclear; however, this is due to a lack of a clearly defined null allele. Here we report the first defined null mutation of Su(dx), generated by P-element excision, which removes the complete open reading frame. We show that the mutation is recessive-viable, with the Notch gain of function phenotypes affecting wing vein and leg development. We further uncover new roles for Su(dx) in Drosophila oogenesis, where it regulates interfollicular stalk formation, egg chamber separation and germline cyst enwrapment by the follicle stem cells. Interestingly, while the null allele exhibited a gain in Notch activity during oogenesis, the previously described Su(dx)SP allele, which carries a seven amino acid in-frame deletion, displayed a Notch loss of function phenotypes and an increase in follicle stem cell turnover. This is despite both alleles displaying similar Notch gain of function in wing development. We attribute this unexpected context-dependent outcome of Su(dx)sp being due to the partial retention of function by the intact C2 and WW domain regions of the protein. Our results extend our understanding of the developmental role of Su(dx) in the tissue renewal and homeostasis of the Drosophila ovary and illustrate the importance of examining an allelic series of mutations to fully understand developmental functions.


Subject(s)
Alleles , Drosophila Proteins , Drosophila melanogaster , Oogenesis , Receptors, Notch , Animals , Oogenesis/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Female , Wings, Animal/growth & development , Wings, Animal/metabolism , Mutation , Signal Transduction , Phenotype , Membrane Proteins
16.
Reprod Biol ; 24(2): 100883, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643607

ABSTRACT

Fibroblast growth factor 10 (FGF10) plays critical roles in oocyte maturation and embryonic development; however, the specific pathway by which FGF10 promotes in vitro maturation of buffalo oocytes remains elusive. The present study was aimed at investigating the mechanism underlying effects of the FGF10-mediated extracellular regulated protein kinases (ERK) pathway on oocyte maturation and embryonic development in vitro. MEK1/2 (mitogen-activated protein kinase kinase) inhibitor U0126, alone or in combination with FGF10, was added to the maturation culture medium during maturation of the cumulus oocyte complex. Morphological observations, orcein staining, apoptosis detection, and quantitative real-time PCR were performed to evaluate oocyte maturation, embryonic development, and gene expression. U0126 affected oocyte maturation and embryonic development in vitro by substantially reducing the nuclear maturation of oocytes and expansion of the cumulus while increasing the apoptosis of cumulus cells. However, it did not have a considerable effect on glucose metabolism. These findings suggest that blocking the MEK/ERK pathway is detrimental to the maturation and embryonic development potential of buffalo oocytes. Overall, FGF10 may regulate the nuclear maturation of oocytes and cumulus cell expansion and apoptosis but not glucose metabolism through the MEK/ERK pathway. Our findings indicate that FGF10 regulates resumption of meiosis and expansion and survival of cumulus cells via MEK/ERK signaling during in vitro maturation of buffalo cumulus oocyte complexes. Elucidation of the mechanism of action of FGF10 and insights into oocyte maturation should advance buffalo breeding. Further studies should examine whether enhancement of MEK/ERK signaling improves embryonic development in buffalo.


Subject(s)
Buffaloes , Butadienes , Fibroblast Growth Factor 10 , In Vitro Oocyte Maturation Techniques , Nitriles , Oocytes , Animals , Buffaloes/embryology , Fibroblast Growth Factor 10/pharmacology , Butadienes/pharmacology , Oocytes/drug effects , In Vitro Oocyte Maturation Techniques/veterinary , Nitriles/pharmacology , Female , Oogenesis/drug effects , Cumulus Cells/drug effects , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Embryonic Development/drug effects , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism
17.
Tissue Cell ; 88: 102372, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38598872

ABSTRACT

Ovarian somatic tissues typically surround developing oocytes and play a crucial role in oogenesis across various metazoans, often displaying structural properties specific to their functions. However, there is an absence of evident structural modularity in the follicle epithelium of Myriapoda. We report here two structurally and developmentally distinct domains within the follicle epithelium of the Japanese pill millipede, Hyleoglomeris japonica. The follicle epithelium of H. japonica exhibits a thick cell mass at the apex of the follicle. These cells harbor abundant rough endoplasmic reticulum, mitochondria, Golgi complexes, and numerous microvilli, indicative of synthetic/secretory activities. Moreover, their height increases as oogenesis progresses. In contrast, another region of the epithelium lacks these features. Our findings highlight the presence of structural and functional modularity in the follicle epithelium of H. japonica. We suggest classifying the follicle epithelium of Myriapoda into three types: homogenous epithelia with enhanced synthetic activities, homogenous epithelia with diminished such activities, and heterogeneous epithelia with varying synthetic activities. These findings prompt a reevaluation of the nature of ovarian somatic tissues in Myriapoda as well as in Arthropoda.


Subject(s)
Arthropods , Ovarian Follicle , Animals , Female , Ovarian Follicle/cytology , Arthropods/ultrastructure , Epithelium , Oogenesis/physiology
18.
J Assist Reprod Genet ; 41(5): 1387-1401, 2024 May.
Article in English | MEDLINE | ID: mdl-38656738

ABSTRACT

OBJECTIVE: Women who are of reproductive age can suffer from polycystic ovary syndrome (PCOS), an endocrine disorder. Anovulatory infertility is mostly caused by aberrant follicular development, which is seen in PCOS patients. Due to the dysfunction of reproductive and endocrine function in PCOS patients, assisted reproduction treatment is one of the main means to obtain clinical pregnancy for PCOS patients. Long non-coding RNA (lncRNA) as a group of functional RNA molecules have been found to participate in the regulation of oocyte function, hormone metabolism, and proliferation and apoptosis of granulosa cells. In this study, we investigated the role of lncRNAs in follicular fluid-derived exosomes and the underlying mechanism of lncRNA LIPE-AS1. METHODS: We used RNA sequencing to analyze the lncRNA profiles of follicular fluid-derived exosomes in PCOS patients and controls. RT-qPCR was performed to detect the expression levels of these lncRNAs in control (n = 30) and PCOS (n = 30) FF exosome samples. Furthermore, we validated the performance of lncRNA LIPE-AS1 in oocyte maturation by in vitro maturation (IVM) experiments in mouse and steroid metabolism in granulosa cells. RESULTS: We found 501 lncRNAs were exclusively expressed in the control group and another 273 lncRNAs were found to be specifically expressed in the PCOS group. LncRNA LIPE-AS1, highly expressed in PCOS exosomes, was related to a poor oocyte maturation and embryo development in PCOS patients. Reduced number of MII oocytes were observed in the LIPE-AS1 group by in vitro maturation (IVM) experiments in mouse. LIPE-AS1 was also shown to modulate steroid metabolism and granulosa cell proliferation and apoptosis by LIPE-AS1/miR-4306/LHCGR axis. CONCLUSION: These findings suggested that the increased expression of LIPE-AS1, facilitated by follicular fluid exosomes, had a significant impact on both oocyte maturation and embryo development. We demonstrated the ceRNA mechanism involving LIPE-AS1, miR-4306, and LHCGR as a regulator of hormone production and metabolism. These findings indicate that LIPE-AS1 is essential in PCOS oocyte maturation and revealed a ceRNA network of LIPE-AS1 and provided new information on abnormal steroid metabolism and oocyte development in PCOS.


Subject(s)
Exosomes , Follicular Fluid , Granulosa Cells , Oocytes , Polycystic Ovary Syndrome , RNA, Long Noncoding , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/metabolism , Female , Follicular Fluid/metabolism , RNA, Long Noncoding/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Exosomes/genetics , Exosomes/metabolism , Oocytes/metabolism , Oocytes/growth & development , Mice , Animals , In Vitro Oocyte Maturation Techniques , Adult , Steroids/metabolism , Oogenesis/genetics , Apoptosis/genetics , Cell Proliferation/genetics
19.
Sci Rep ; 14(1): 8263, 2024 04 09.
Article in English | MEDLINE | ID: mdl-38594333

ABSTRACT

Oocytes of both vertebrates and invertebrates often contain an intricate organelle assemblage, termed the Balbiani body (Bb). It has previously been suggested that this assemblage is involved in the delivery of organelles and macromolecules to the germ plasm, formation of oocyte reserve materials, and transfer of mitochondria to the next generation. To gain further insight into the function of the Bb, we performed a series of analyses and experiments, including computer-aided 3-dimensional reconstructions, detection of DNA (mtDNA) synthesis as well as immunolocalization studies. We showed that in orthopteran Meconema meridionale, the Bb comprises a network of mitochondria and perinuclear nuage aggregations. As oogenesis progresses, the network expands filling almost entire ooplasm, then partitions into several smaller entities, termed micro-networks, and ultimately into individual mitochondria. As in somatic cells, this process involves microfilaments and elements of endoplasmic reticulum. We showed also that at least some of the individual mitochondria are surrounded by phagophores and eliminated via mitophagy. These findings support the idea that the Bb is implicated in the multiplication and selective elimination of (defective) mitochondria and therefore may participate in the transfer of undamaged (healthy) mitochondria to the next generation.


Subject(s)
Oocytes , Orthoptera , Animals , Oocytes/metabolism , Oogenesis/genetics , Mitochondria/genetics , Insecta , Endoplasmic Reticulum
20.
Nutr Diabetes ; 14(1): 23, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653987

ABSTRACT

BACKGROUND: The number of patients with type 1 diabetes rises rapidly around the world in recent years. Maternal diabetes has a detrimental effect on reproductive outcomes due to decreased oocyte quality. However, the strategies to improve the oocyte quality and artificial reproductive technology (ART) efficiency of infertile females suffering from diabetes have not been fully studied. In this study, we aimed to examine the effects of nicotinamide mononucleotide (NMN) on oocyte maturation of mouse with type 1 diabetes mouse and explore the underlying mechanisms of NMN's effect. METHODS: Streptozotocin (STZ) was used to establish the mouse models with type 1 diabetes. The successful establishment of the models was confirmed by the results of body weight test, fasting blood glucose test and haematoxylin and eosin (H&E) staining. The in vitro maturation (IVM) rate of oocytes from diabetic mice was examined. Immunofluorescence staining (IF) was performed to examine the reactive oxygen species (ROS) level, spindle/chromosome structure, mitochondrial function, actin dynamics, DNA damage and histone modification of oocytes, which are potential factors affecting the oocyte quality. The quantitative reverse transcription PCR (RT-qPCR) was used to detect the mRNA levels of Sod1, Opa1, Mfn2, Drp1, Sirt1 and Sirt3 in oocytes. RESULTS: The NMN supplementation increased the oocyte maturation rate of the mice with diabetes. Furthermore, NMN supplementation improved the oocyte quality by rescuing the actin dynamics, reversing meiotic defects, improving the mitochondrial function, reducing ROS level, suppressing DNA damage and restoring changes in histone modifications of oocytes collected from the mice with diabetes. CONCLUSION: NMN could improve the maturation rate and quality of oocytes in STZ-induced diabetic mice, which provides a significant clue for the treatment of infertility of the patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Dynamins , Nicotinamide Mononucleotide , Oocytes , Reactive Oxygen Species , Animals , Mice , Female , Oocytes/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Experimental/drug therapy , Reactive Oxygen Species/metabolism , Nicotinamide Mononucleotide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Sirtuin 1/metabolism , Sirtuin 3/metabolism , In Vitro Oocyte Maturation Techniques/methods , Superoxide Dismutase-1 , DNA Damage/drug effects , Streptozocin , Oogenesis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...