Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52.567
Filtrer
1.
J Ovarian Res ; 17(1): 137, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961417

RÉSUMÉ

BACKGROUND: The utilization of a double trigger, involving the co-administration of gonadotropin-releasing hormone agonist (GnRH-a) and human chorionic gonadotropin (hCG) for final oocyte maturation, is emerging as a novel approach in gonadotropin-releasing hormone antagonist (GnRH-ant) protocols during controlled ovarian hyperstimulation (COH). This protocol involves administering GnRH-a and hCG 40 and 34 h prior to ovum pick-up (OPU), respectively. This treatment modality has been implemented in patients with low/poor oocytes yield. This study aimed to determine whether the double trigger could improve the number of top-quality embryos (TQEs) in patients with fewer than three TQEs. METHODS: The stimulation characteristics of 35 in vitro fertilization (IVF) cycles were analyzed. These cycles were triggered by the combination of hCG and GnRHa (double trigger cycles) and compared to the same patients' previous IVF attempt, which utilized the hCG trigger (hCG trigger control cycles). The analysis involved cases who were admitted to our reproductive center between January 2018 and December 2022. In the hCG trigger control cycles, all 35 patients had fewer than three TQEs. RESULTS: Patients who received the double trigger cycles yielded a significantly higher number of 2PN cleavage embryos (3.54 ± 3.37 vs. 2.11 ± 2.15, P = 0.025), TQEs ( 2.23 ± 2.05 vs. 0.89 ± 0.99, P < 0.001), and a simultaneously higher proportion of the number of cleavage stage embryos (53.87% ± 31.38% vs. 39.80% ± 29.60%, P = 0.043), 2PN cleavage stage embryos (43.89% ± 33.01% vs. 27.22% ± 27.13%, P = 0.014), and TQEs (27.05% ± 26.26% vs. 14.19% ± 19.76%, P = 0.019) to the number of oocytes retrieved compared with the hCG trigger control cycles, respectively. The double trigger cycles achieved higher rates of cumulative clinical pregnancy (20.00% vs. 2.86%, P = 0.031), cumulative persistent pregnancy (14.29% vs. 0%, P < 0.001), and cumulative live birth (14.29% vs. 0%, P < 0.001) per stimulation cycle compared with the hCG trigger control cycles. CONCLUSION: Co-administration of GnRH-agonist and hCG for final oocyte maturation, 40 and 34 h prior to OPU, respectively (double trigger) may be suggested as a valuable new regimen for treating patients with low TQE yield in previous hCG trigger IVF/intracytoplasmic sperm injection (ICSI) cycles.


Sujet(s)
Gonadotrophine chorionique , Fécondation in vitro , Hormone de libération des gonadotrophines , Ovocytes , Induction d'ovulation , Humains , Femelle , Gonadotrophine chorionique/administration et posologie , Gonadotrophine chorionique/usage thérapeutique , Hormone de libération des gonadotrophines/agonistes , Hormone de libération des gonadotrophines/antagonistes et inhibiteurs , Adulte , Fécondation in vitro/méthodes , Induction d'ovulation/méthodes , Grossesse , Ovocytes/effets des médicaments et des substances chimiques , Injections intracytoplasmiques de spermatozoïdes/méthodes , Taux de grossesse , Ovogenèse/effets des médicaments et des substances chimiques
2.
Am J Reprod Immunol ; 92(1): e13902, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39042556

RÉSUMÉ

PROBLEM: Lipopolysaccharide (LPS) from gram-negative bacteria has reportedly been associated with infectious diseases like metritis, which has a substantial adverse effect on animal reproductive performance and causes serious financial losses for the dairy sector. The current work aimed to establish the impact of LPS on in vitro oocyte maturation and subsequent in vitro developmental competence of oocytes, as well as to investigate the explanatory molecular mechanism underlying this effect. METHOD OF STUDY: Buffalo cumulus-oocyte complexes (COCs) were challenged with 0, 5, 10 and 20 µg/mL LPS during IVM followed by IVF and IVC. Cytoplasmic and nuclear maturation, cleavage and blastocyst rate, intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP, ΔΨm) and transcript abundance of genes related to inflammation, antioxidation and apoptosis were evaluated. RESULTS: The maturation and subsequent embryonic development competency were found to be significantly (p ≤ 0.05) reduced with the addition of 10 and 20 µg/mL LPS to IVM media. ROS production accompanied by a decreased ΔΨm was recorded in LPS-treated oocytes in comparison to the control group (p ≤ 0.05). Our results were further supported by the transcriptional expression of proinflammatory (TLR4, CD14 and RPS27A) and apoptotic gene (Caspase 3) which were found to be significantly increased while antioxidant genes (SOD2 and GPX1) were decreased significantly in matured oocytes and blastocyst after LPS exposure. CONCLUSIONS: The deleterious effects of LPS are mediated through ROS generation, which triggers inflammatory processes via the TLR4 pathway and impairs oocyte maturation and subsequent embryonic development.


Sujet(s)
Buffles , Développement embryonnaire , Techniques de maturation in vitro des ovocytes , Lipopolysaccharides , Mitochondries , Ovocytes , Espèces réactives de l'oxygène , Transduction du signal , Récepteur de type Toll-4 , Animaux , Récepteur de type Toll-4/métabolisme , Espèces réactives de l'oxygène/métabolisme , Ovocytes/métabolisme , Ovocytes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Développement embryonnaire/effets des médicaments et des substances chimiques , Femelle , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Cellules cultivées , Blastocyste/métabolisme , Blastocyste/effets des médicaments et des substances chimiques , Fécondation in vitro
3.
Anticancer Res ; 44(8): 3577-3586, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060058

RÉSUMÉ

BACKGROUND/AIM: Age-related decline in the number of ovulations and ovum quality are major causes of female infertility, and stem cells have been reported to be effective in tissue regeneration. However, current therapeutic modalities are inadequate. This study investigated the effects of adipose-derived mesenchymal stem cells (ASCs) on ovarian functions in aged mice. MATERIALS AND METHODS: Following the characterization of ASCs using flow cytometry, the effects of ASCs on the number of ovulations, fertilization rate, and blastocyst-formation rate were investigated. In addition, the number of ovarian follicles and serum anti-Müllerian hormone (AMH) levels were examined. ASCs marked with Kusabira Orange were used to examine the location after cell administration. The quality of ovulated oocytes was analyzed using next-generation RNA sequencing. RESULTS: ASCs showed characteristics of mesenchymal stem cells and were distributed to various organs, including the ovarian stroma. The transplantation resulted in increased number of oocytes and ovulation in the ovaries and increased AMH values. Genetic analysis revealed improved oocyte quality and increased fertilization and blastocyst-formation rates. CONCLUSION: ASC therapy may be effective in improving fertility in older women.


Sujet(s)
Tissu adipeux , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Ovocytes , Ovaire , Animaux , Femelle , Transplantation de cellules souches mésenchymateuses/méthodes , Souris , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Tissu adipeux/cytologie , Vieillissement/physiologie , Hormone antimullérienne/sang , Hormone antimullérienne/métabolisme , Follicule ovarique/métabolisme , Follicule ovarique/cytologie , Ovulation
4.
Nat Commun ; 15(1): 6323, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39060312

RÉSUMÉ

The timing of DNA replication in mammals is crucial for minimizing errors and influenced by genome usage and chromatin states. Replication timing in the newly formed mammalian embryo remains poorly understood. Here, we have investigated replication timing in mouse zygotes and 2-cell embryos, revealing that zygotes lack a conventional replication timing program, which then emerges in 2-cell embryos. This program differs from embryonic stem cells and generally correlates with transcription and genome compartmentalization of both parental genomes. However, consistent and systematic differences existed between the replication timing of the two parental genomes, including considerably later replication of maternal pericentromeric regions compared to paternal counterparts. Moreover, maternal chromatin modified by Polycomb Repressive Complexes in the oocyte, undergoes early replication, despite belonging to the typically late-replicating B-compartment of the genome. This atypical and asynchronous replication of the two parental genomes may advance our understanding of replication stress in early human embryos and trigger strategies to reduce errors and aneuploidies.


Sujet(s)
Réplication de l'ADN , Embryon de mammifère , Zygote , Animaux , Femelle , Souris , Zygote/métabolisme , Mâle , Embryon de mammifère/métabolisme , Chromatine/métabolisme , Chromatine/génétique , Ovocytes/métabolisme , Déroulement de la réplication de l'ADN , Génome , Développement embryonnaire/génétique , Souris de lignée C57BL
5.
Sci Rep ; 14(1): 17183, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39060327

RÉSUMÉ

Pre-incubation of the cumulus-oocyte complex (COCs) may lead to better function of cumulus cells (CCs) and higher oocyte quality by changing the transcriptomic profile of CCs. 140 cumulus cell samples were isolated from 12 participants and divided into two groups based on pre-incubation time. In the T0 group, the COCs were immediately dissected to separate the CCs from around the oocytes. In the T2 group, CCs were prepared after 2 h of incubation. Then, the transcriptomic profile of the CCs of the non pre-incubation group was compared to the 2-h pre-incubation group. Confirmation of RNA sequencing results was done via qRT­PCR. The CCs transcriptome analysis showed 17 genes were downregulated and 22 genes upregulated in the T2 group compared to the T0 group. Also, the pathways related to ATP production (oxidative phosphorylation, electron transport chain, and Mitochondrial complex I assembly model OXPHOS system), TNF-alpha signaling pathway, and glucocorticoid receptor pathway increased in the T2 group compared to the T0 group. Also, the TGF-ß pathway was decreased in the T2 group compared to the T0 group. This study showed that 2 h pre-incubation leads to changes in important pathways in CCs, which positively affects oocyte quality.


Sujet(s)
Cellules du cumulus , Ovocytes , Transcriptome , Cellules du cumulus/métabolisme , Humains , Femelle , Ovocytes/métabolisme , Adulte , Analyse de profil d'expression de gènes , Phosphorylation oxydative , Transduction du signal
6.
J Cell Mol Med ; 28(12): e18487, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39031722

RÉSUMÉ

Premature ovarian insufficiency (POI) is one of the important causes of female infertility. Yet the aetiology for POI is still elusive. FBXW7 (F-box with 7 tandem WD) is one of the important components of the Skp1-Cullin1-F-box (SCF) E3 ubiquitin ligase. FBXW7 can regulate cell growth, survival and pluripotency through mediating ubiquitylation and degradation of target proteins via triggering the ubiquitin-proteasome system, and is associated with tumorigenesis, haematopoiesis and testis development. However, evidence establishing the function of FBXW7 in ovary is still lacking. Here, we showed that FBXW7 protein level was significantly decreased in the ovaries of the cisplatin-induced POI mouse model. We further showed that mice with oocyte-specific deletion of Fbxw7 demonstrated POI, characterized with folliculogenic defects, early depletion of follicle reserve, disordered hormonal secretion, ovarian dysfunction and female infertility. Impaired oocyte-GCs communication, manifested as down-regulation of connexin 37, may contribute to follicular development failure in the Fbxw7-mutant mice. Furthermore, single-cell RNA sequencing and in situ hybridization results indicated an accumulation of Clu and Ccl2 transcripts, which may alter follicle microenvironment deleterious to oocyte development and accelerate POI. Our results establish the important role of Fbxw7 in folliculogenesis and ovarian function, and might provide valuable information for understanding POI and female infertility.


Sujet(s)
Protéine-7 contenant une boite F et des répétitions WD , Ovocytes , Follicule ovarique , Insuffisance ovarienne primitive , Animaux , Femelle , Insuffisance ovarienne primitive/génétique , Insuffisance ovarienne primitive/métabolisme , Insuffisance ovarienne primitive/anatomopathologie , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Ovocytes/métabolisme , Souris , Follicule ovarique/métabolisme , Follicule ovarique/croissance et développement , Follicule ovarique/anatomopathologie , Modèles animaux de maladie humaine , Délétion de gène , Souris knockout , Infertilité féminine/génétique , Infertilité féminine/métabolisme , Infertilité féminine/anatomopathologie , Cisplatine/effets indésirables
7.
FASEB J ; 38(14): e23837, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39031536

RÉSUMÉ

Bone morphogenetic protein 15 (BMP15) is an oocyte-specific growth factor important for successful female reproduction in mammals. While mutations in BMP15/Bmp15 cause ovulatory deficiency and/or infertility in certain mammalian species, loss of bmp15 in zebrafish, a continuous spawner and the only bmp15 knockout model in fish to date, results in complete arrest of follicle development and later female-to-male sex reversal, preventing to examine effects on ovulation/fertilization. Here, we used Atlantic salmon, a seasonal spawner, and generated bmp15 mutants to investigate ovarian development and fertility. Histological and morphometric analyses revealed that in biallelic frameshift (bmp15 fs/fs) mutant ovaries, folliculogenesis started earlier, resulting in an advanced development compared to wild-type (WT) controls, accompanied by a weaker expression of the (early) oocyte-specific factor figla. This precocious ovarian development was followed in bmp15 fs/fs females by enhanced follicle atresia during vitellogenic stages. Although genes involved in steroid synthesis and signaling (star, cyp11b, cyp17a1 and esr1) were dramatically higher in late vitellogenic bmp15 fs/fs mutant ovaries, estradiol-17ß plasma levels were lower than in WT counterparts, potentially reflecting compensatory changes at the level of ovarian gene expression. At spawning, bmp15 fs/fs females displayed lower gonado-somatic index values and reduced oocyte diameter, and the majority (71.4%), showed mature non-ovulating ovaries with a high degree of atresia. The remaining (28.6%) females spawned eggs but they either could not be fertilized or, upon fertilization, showed severe malformations and embryonic mortality. Our results show that Bmp15 is required for proper follicle recruitment and growth and later ovulatory success in Atlantic salmon, providing an alternative candidate target to induce sterility in farmed salmon. Moreover, since loss of bmp15 in salmon, in contrast to zebrafish, does not result in female-to-male sex change, this is the first mutant model in fish allowing further investigations on Bmp15-mediated functions in the ovulatory period.


Sujet(s)
Protéine morphogénétique osseuse de type 15 , Ovulation , Salmo salar , Animaux , Protéine morphogénétique osseuse de type 15/génétique , Protéine morphogénétique osseuse de type 15/métabolisme , Femelle , Salmo salar/métabolisme , Salmo salar/génétique , Salmo salar/croissance et développement , Ovaire/métabolisme , Follicule ovarique/métabolisme , Ovocytes/métabolisme , Mâle , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Saisons
8.
Theriogenology ; 226: 253-262, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38950486

RÉSUMÉ

The potential of using long in vitro culture (LIVC) of cumulus-oocyte complexes (COCs) from early antral follicles (EAFs) as an assisted reproductive technology in cattle has shown promising results. This study explored the feasibility of applying this technology to sheep as seasonal breeding animals. Ovaries from sheep were collected during both the breeding and non-breeding seasons. COCs were isolated from EAFs (350-450 µm) and cultured in TCM199 medium supplemented with 0.15 µg/mL Zn sulfate, 10-4IU/mL FSH, 10 ng/mL estradiol, 50 ng/mL testosterone, 50 ng/mL progesterone, and 5 µM Cilostamide. After five days of LIVC, the COCs were submitted to an in vitro maturation procedure. The results indicate successful in vitro development of COCs, evidenced by a significant increase in oocyte diameter (p < 0.000) and the preservation of gap junction communication between oocyte and cumulus cells. The gradual uncoupling was accompanied by a progressive chromatin transition from the non-surrounded nucleolus (NSN) to the surrounded nucleolus (SN) (p < 0.000), coupled with a gradual decrease in global transcriptional activity and an increase in oocyte meiotic competence (p < 0.000). Maintenance of oocyte-cumulus investment architecture, viability, and metaphase II capability was significantly higher in COCs collected during the breeding season (p < 0.000), suggesting higher quality than those obtained during the non-breeding season. In conclusion, our study confirms LIVC feasibility in sheep, emphasizing increased effectiveness during the breeding season in isolating higher-quality COCs from EAFs. These findings can influence improving the LIVC system in mammals with seasonal reproduction.


Sujet(s)
Techniques de maturation in vitro des ovocytes , Méiose , Ovocytes , Follicule ovarique , Animaux , Ovis/physiologie , Femelle , Ovocytes/physiologie , Techniques de maturation in vitro des ovocytes/médecine vétérinaire , Techniques de maturation in vitro des ovocytes/méthodes , Follicule ovarique/physiologie , Cellules du cumulus/physiologie
9.
Theriogenology ; 226: 286-293, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38954997

RÉSUMÉ

HT-2 toxin is a type of mycotoxin which is shown to affect gastric and intestinal lesions, hematopoietic and immunosuppressive effects, anorexia, lethargy, nausea. Recently, emerging evidences indicate that HT-2 also disturbs the reproductive system. In this study, we investigated the impact of HT-2 toxin exposure on the organelles of porcine oocytes. Our results found that the abnormal distribution of endoplasmic reticulum increased after HT-2 treatment, with the perturbation of ribosome protein RPS3 and GRP78 expression; Golgi apparatus showed diffused localization pattern and GM130 localization was also impaired, thereby affecting the Rab10-based vesicular transport; Due to the impairment of ribosomes, ER, and Golgi apparatus, the protein supply to lysosomes was hindered, resulting in lysosomal damage, which further disrupted the LC3-based autophagy. Moreover, the results indicated that the function and distribution of mitochondria were also affected by HT-2 toxin, showing with fragments of mitochondria, decreased TMRE and ATP level. Taken together, our study suggested that HT-2 toxin exposure induces damage to the organelles for endomembrane system, which further inhibited the meiotic maturation of porcine oocytes.


Sujet(s)
Techniques de maturation in vitro des ovocytes , Ovocytes , Animaux , Suidae , Ovocytes/effets des médicaments et des substances chimiques , Techniques de maturation in vitro des ovocytes/médecine vétérinaire , Toxine T-2/toxicité , Toxine T-2/analogues et dérivés , Femelle , Appareil de Golgi/effets des médicaments et des substances chimiques , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques
10.
Theriogenology ; 226: 335-342, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38959844

RÉSUMÉ

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.


Sujet(s)
Méiose , Mitogen-Activated Protein Kinase 7 , Ovocytes , Appareil du fuseau , Animaux , Ovocytes/physiologie , Méiose/physiologie , Souris , Appareil du fuseau/physiologie , Mitogen-Activated Protein Kinase 7/métabolisme , Mitogen-Activated Protein Kinase 7/génétique , Femelle
11.
Curr Biol ; 34(13): R628-R630, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38981428

RÉSUMÉ

Meiotic cohesion loss underlies elevated rates of infertility and chromosome abnormalities in children of older women. A new study shows that cohesins are turned over throughout meiotic prophase, suggesting that cohesion loss is likely not solely due to early establishment of cohesion.


Sujet(s)
Protéines chromosomiques nonhistones , Ovocytes , Ovocytes/physiologie , Femelle , Humains , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Méiose , Animaux
12.
Syst Biol Reprod Med ; 70(1): 195-203, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38972054

RÉSUMÉ

The presence of cyclic adenosine monophosphate (cAMP) has been considered to be a fundamental factor in ensuring meiotic arrest prior to ovulation. cAMP is regarded as a key molecule in the regulation of oocyte maturation. However, it has been reported that increased levels of intracellular cAMP can result in abnormal cytokinesis, with some MI oocytes leading to symmetrically cleaved 2-cell MII oocytes. Consequently, we aimed to investigate the effects of elevated intracellular cAMP levels on abnormal cytokinesis and oocyte maturation during the meiosis of mouse oocytes. This study found that a high concentration of isobutylmethylxanthine (IBMX) also caused chromatin/chromosomes aggregation (AC) after the first meiosis. The rates of AC increased the greater the concentration of IBMX. In addition, AC formation was found to be reversible, showing that the re-formation of the spindle chromosome complex was possible after the IBMX was removed. In human oocytes, the chromosomes aggregate after the germinal vesicle breakdown and following the first and second polar body extrusions (the AC phase), while mouse oocytes do not have this AC phase. The results of our current study may indicate that the AC phase in human oocytes could be related to elevated levels of intracytoplasmic cAMP.


Sujet(s)
Xanthine(isobutyl-3 methyl-1) , Chromatine , Ovocytes , Animaux , Ovocytes/métabolisme , Femelle , Chromatine/métabolisme , Xanthine(isobutyl-3 methyl-1)/pharmacologie , Souris , Humains , Méiose/effets des médicaments et des substances chimiques , AMP cyclique/métabolisme , Inhibiteurs de la phosphodiestérase/pharmacologie
13.
J Gen Physiol ; 156(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38968404

RÉSUMÉ

We used voltage clamp fluorometry to probe the movement of the S4 helix in the voltage-sensing domain of the sea urchin HCN channel (spHCN) expressed in Xenopus oocytes. We obtained markedly different fluorescence responses with either ALEXA-488 or MTS-TAMRA covalently linked to N-terminal Cys332 of the S4 helix. With hyperpolarizing steps, ALEXA-488 fluorescence increased rapidly, consistent with it reporting the initial inward movement of S4, as previously described. In contrast, MTS-TAMRA fluorescence increased more slowly and its early phase correlated with that of channel opening. Additionally, a slow fluorescence component that tracked the development of the mode shift, or channel hysteresis, could be resolved with both labels. We quantitated this component as an increased deactivation tail current delay with concomitantly longer activation periods and found it to depend strongly on the presence of K+ ions in the pore. Using collisional quenching experiments and structural predictions, we established that ALEXA-488 was more exposed to solvent than MTS-TAMRA. We propose that components of S4 movement during channel activation can be kinetically resolved using different fluorescent probes to reveal distinct biophysical properties. Our findings underscore the need to apply caution when interpreting voltage clamp fluorometry data and demonstrate the potential utility of different labels to interrogate distinct biophysical properties of voltage-gated membrane proteins.


Sujet(s)
Colorants fluorescents , Xenopus laevis , Animaux , Colorants fluorescents/composition chimique , Ouverture et fermeture des portes des canaux ioniques/physiologie , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/métabolisme , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/génétique , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/composition chimique , Ovocytes/métabolisme , Echinoidea , Potentiels de membrane/physiologie
14.
Science ; 385(6704): 15, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38963833

RÉSUMÉ

Studies find long-lived proteins are prevalent in the organs.


Sujet(s)
Ovocytes , Ovaire , Protéines , Animaux , Femelle , Humains , Souris , Ovaire/métabolisme , Protéines/métabolisme , Protéines/composition chimique , Ovocytes/métabolisme
15.
J Ovarian Res ; 17(1): 148, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39020390

RÉSUMÉ

Vitamin D3 plays a crucial role in female reproduction. As research progresses, the mechanisms of action of vitamin D3 on follicular development have been widely discussed. Firstly, key enzymes involved in the synthesis and metabolism of vitamin D3 have been discovered in the ovary, suggesting that vitamin D3 can be synthesized and metabolized locally within the ovary. Additionally, the detection of vitamin D3 receptors (VDR) in follicles suggests that vitamin D3 may exert its effects by binding specifically to these receptors during follicular development. Further research indicates that vitamin D3 promotes follicular growth by enhancing the development of granulosa cells (GCs) and oocytes. Currently, the mechanism of action of vitamin D3 in follicular development is becoming increasingly clear. Vitamin D3 promotes oocyte development by regulating molecules involved in meiotic arrest in oocytes. It also enhances granulosa cell proliferation by stimulating steroid hormone synthesis and cell cycle regulation. Additionally, vitamin D3 exerts anti-inflammatory effects by reducing oxidative stress and advanced glycation end-products (AGEs), mitigating the detrimental effects of inflammation on follicular development. These functions of vitamin D3 have clinical applications, such as in treating polycystic ovary syndrome (PCOS), improving female fertility, and enhancing outcomes in in vitro fertilization (IVF). This review summarizes the research progress on the role and mechanisms of vitamin D3 in follicular development and briefly summarizes its clinical applications.


Sujet(s)
Cholécalciférol , Follicule ovarique , Humains , Femelle , Cholécalciférol/métabolisme , Follicule ovarique/métabolisme , Animaux , Ovocytes/métabolisme , Cellules de la granulosa/métabolisme , Récepteur calcitriol/métabolisme
16.
Science ; 385(6706): eadn5529, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39024439

RÉSUMÉ

Meiotic errors of relatively small chromosomes in oocytes result in egg aneuploidies that cause miscarriages and congenital diseases. Unlike somatic cells, which preferentially mis-segregate larger chromosomes, aged oocytes preferentially mis-segregate smaller chromosomes through unclear processes. Here, we provide a comprehensive three-dimensional chromosome identifying-and-tracking dataset throughout meiosis I in live mouse oocytes. This analysis reveals a prometaphase pathway that actively moves smaller chromosomes to the inner region of the metaphase plate. In the inner region, chromosomes are pulled by stronger bipolar microtubule forces, which facilitates premature chromosome separation, a major cause of segregation errors in aged oocytes. This study reveals a spatial pathway that facilitates aneuploidy of small chromosomes preferentially in aged eggs and implicates the role of the M phase in creating a chromosome size-based spatial arrangement.


Sujet(s)
Aneuploïdie , Ségrégation des chromosomes , Méiose , Microtubules , Ovocytes , Animaux , Femelle , Souris , Chromosomes de mammifère/génétique , Métaphase , Microtubules/métabolisme , Ovocytes/cytologie , Ovocytes/métabolisme , Jeux de données comme sujet
17.
Proc Natl Acad Sci U S A ; 121(28): e2315043121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968128

RÉSUMÉ

Only 30% of embryos from in vitro fertilized oocytes successfully implant and develop to term, leading to repeated transfer cycles. To reduce time-to-pregnancy and stress for patients, there is a need for a diagnostic tool to better select embryos and oocytes based on their physiology. The current standard employs brightfield imaging, which provides limited physiological information. Here, we introduce METAPHOR: Metabolic Evaluation through Phasor-based Hyperspectral Imaging and Organelle Recognition. This non-invasive, label-free imaging method combines two-photon illumination and AI to deliver the metabolic profile of embryos and oocytes based on intrinsic autofluorescence signals. We used it to classify i) mouse blastocysts cultured under standard conditions or with depletion of selected metabolites (glucose, pyruvate, lactate); and ii) oocytes from young and old mouse females, or in vitro-aged oocytes. The imaging process was safe for blastocysts and oocytes. The METAPHOR classification of control vs. metabolites-depleted embryos reached an area under the ROC curve (AUC) of 93.7%, compared to 51% achieved for human grading using brightfield imaging. The binary classification of young vs. old/in vitro-aged oocytes and their blastulation prediction using METAPHOR reached an AUC of 96.2% and 82.2%, respectively. Finally, organelle recognition and segmentation based on the flavin adenine dinucleotide signal revealed that quantification of mitochondria size and distribution can be used as a biomarker to classify oocytes and embryos. The performance and safety of the method highlight the accuracy of noninvasive metabolic imaging as a complementary approach to evaluate oocytes and embryos based on their physiology.


Sujet(s)
Blastocyste , Ovocytes , Animaux , Blastocyste/métabolisme , Souris , Ovocytes/métabolisme , Femelle , Organites/métabolisme , Imagerie optique/méthodes
18.
Adv Anat Embryol Cell Biol ; 238: 23-46, 2024.
Article de Anglais | MEDLINE | ID: mdl-39030353

RÉSUMÉ

The regulation of mRNA transcription and translation is uncoupled during oogenesis. The reason for this uncoupling is two-fold. Chromatin is only accessible to the transcriptional machinery during the growth phase as it condenses prior to resumption of meiosis to ensure faithful segregation of chromosomes during meiotic maturation. Thus, transcription rates are high during this time period in order to produce all of the transcripts needed for meiosis, fertilization, and embryo cleavage until the newly formed embryonic genome becomes transcriptionally active. To ensure appropriate timing of key developmental milestones including chromatin condensation, resumption of meiosis, segregation of chromosomes, and polar body extrusion, the translation of protein from transcripts synthesized during oocyte growth must be temporally regulated. This is achieved by the regulation of mRNA interaction with RNA binding proteins and shortening and lengthening of the poly(A) tail. This chapter details the essential factors that regulate the dynamic changes in mRNA synthesis, storage, translation, and degradation during oocyte growth and maturation.


Sujet(s)
Ovocytes , Ovogenèse , ARN messager , Ovocytes/métabolisme , Animaux , ARN messager/métabolisme , ARN messager/génétique , Ovogenèse/génétique , Ovogenèse/physiologie , Humains , Régulation de l'expression des gènes au cours du développement , Femelle , Méiose , Biosynthèse des protéines
19.
Adv Anat Embryol Cell Biol ; 238: 47-68, 2024.
Article de Anglais | MEDLINE | ID: mdl-39030354

RÉSUMÉ

DNA damage poses a significant challenge to all eukaryotic cells, leading to mutagenesis, genome instability and senescence. In somatic cells, the failure to repair damaged DNA can lead to cancer development, whereas, in oocytes, it can lead to ovarian dysfunction and infertility. The response of the cell to DNA damage entails a series of sequential and orchestrated events including sensing the DNA damage, activating DNA damage checkpoint, chromatin-related conformational changes, activating the DNA damage repair machinery and/or initiating the apoptotic cascade. This chapter focuses on how somatic cells and mammalian oocytes respond to DNA damage. Specifically, we will discuss how and why fully grown mammalian oocytes differ drastically from somatic cells and growing oocytes in their response to DNA damage.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Ovocytes , Ovocytes/métabolisme , Animaux , Humains , Femelle , Mammifères , Apoptose/génétique
20.
Adv Anat Embryol Cell Biol ; 238: 1-22, 2024.
Article de Anglais | MEDLINE | ID: mdl-39030352

RÉSUMÉ

Successful reproduction relies on the union of a single chromosomally normal egg and sperm. Chromosomally normal eggs develop from precursor cells, called oocytes, that have undergone accurate chromosome segregation. The process of chromosome segregation is governed by the oocyte spindle, a unique cytoskeletal machine that splits chromatin content of the meiotically dividing oocyte. The oocyte spindle develops and functions in an idiosyncratic process, which is vulnerable to genetic variation in spindle-associated proteins. Human genetic variants in several spindle-associated proteins are associated with poor clinical fertility outcomes, suggesting that heritable etiologies for oocyte dysfunction leading to infertility exist and that the spindle is a crux for female fertility. This chapter examines the mammalian oocyte spindle through the lens of human genetic variation, covering the genes TUBB8, TACC3, CEP120, AURKA, AURKC, AURKB, BUB1B, and CDC20. Specifically, it explores how patient-identified variants perturb spindle development and function, and it links these molecular changes in the oocyte to their cognate clinical consequences, such as oocyte maturation arrest, elevated egg aneuploidy, primary ovarian insufficiency, and recurrent pregnancy loss. This discussion demonstrates that small genetic errors in oocyte meiosis can result in remarkably far-ranging embryonic consequences, and thus reveals the importance of the oocyte's fine machinery in sustaining life.


Sujet(s)
Ovocytes , Appareil du fuseau , Ovocytes/métabolisme , Humains , Appareil du fuseau/métabolisme , Femelle , Méiose/génétique , Variation génétique , Infertilité féminine/génétique , Animaux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE