Open Access

Oviduct epithelial cell‑derived extracellular vesicles promote the developmental competence of IVF porcine embryos

  • This article is part of the special Issue: Molecular mammalian reproduction
  • Authors:
    • Xun Fang
    • Seonggyu Bang
    • Bereket Molla Tanga
    • Chaerim Seo
    • Dongjie Zhou
    • Gyeonghwan Seong
    • Islam M. Saadeldin
    • Sanghoon Lee
    • Xiang-Shun Cui
    • Jongki Cho
  • View Affiliations

  • Published online on: May 12, 2023     https://doi.org/10.3892/mmr.2023.13009
  • Article Number: 122
  • Copyright: © Fang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Assisted reproductive technology has increased the efficiency of animal reproduction. However, polyspermy is a significant limitation of porcine in vitro fertilization (IVF). Therefore, reducing the polyspermy rate and improving monospermic embryos is crucial. Recent studies have reported that oviductal fluid, along with its contents of extracellular vesicles (EVs), enhanced the fertilization process and supported embryo development. Consequently, the present study investigated the effects of porcine oviduct epithelial cells (OEC‑EVs) on sperm‑oocyte interactions during porcine IVF and evaluated in vitro embryo developmental competence outcomes. During IVF embryo development, the cleavage rate was significantly higher in the group treated with 50 ng/ml OEC‑EVs compared with the control group (67.6±2.5 vs. 57.3±1.9; P<0.05). Furthermore, the OEC‑EV group had significantly more embryos (16.4±1.2 vs. 10.2±0.8; P<0.05), and the polyspermy rate significantly decreased (32.9±2.5 vs. 43.8±3.1; P<0.05) compared with that of the control group. Additionally, the fluorescence intensities of cortical granules (3.56±0.47 vs. 2.15±0.24; P<0.05) and active mitochondria (8.14±0.34 vs. 5.96±0.38; P<0.05) were significantly higher in the OEC‑EV group compared with those in the control group. In conclusion, OEC‑EV adsorption and penetration crosstalk between sperm and oocytes was observed. OEC‑EV treatment was demonstrated to significantly improve the concentration and distribution of cortical granules in oocytes. Furthermore, OEC‑EVs also increased oocyte mitochondrial activity, reduced polyspermy and increased the IVF success rate.

Introduction

In vitro embryo production in pigs is a crucial tool for the development of porcine models suitable for human biomedical research due to their anatomical and physiological similarities to human organs (1). As a result, xenotransplantation using pig tissues and organs is feasible (2,3). Assisted reproductive technology has improved animal reproduction efficiency. However, polyspermy is a major limitation of porcine in vitro fertilization (IVF), which leads to chromosomal abnormalities in embryos (4,5). Hence, reducing the polyspermy rate and enhancing monospermic embryos are crucial for successful porcine IVF.

Standard IVF systems utilize hundreds of spermatozoa, often resulting in a rise in the number of spermatozoa that penetrate the oocyte (6). Polyspermy is strongly linked to the initial number of capacitated spermatozoa during the IVF process (7). One strategy to decrease the incidence of polyspermy is to reduce the absolute number of spermatozoa, but this results in a low oocyte penetration rate. Consequently, certain IVF conditions which mimic the oviductal environment have been examined to minimize polyspermy in vitro, including shortening the co-incubation time and manipulating the IVF equipment or culture conditions (8). These studies have effectively reduced the incidence of polyspermy. However, the available IVF systems do not entirely mimic in vivo conditions (9), and the percentage of monospermic oocytes relative to the total number inseminated during the IVF remains between 20–30% (68).

In mammals, the oviduct is composed of four parts; the infundibulum, responsible for collecting ovulated oocytes; the ampulla, where fertilization occurs; the isthmus, for preimplantation embryo development; and the uterotubal junction, which transits the embryos to the uterus (10). Oviductal fluid (OF) and extracellular vesicles (EVs) secreted by the oviduct epithelial cells (OEC; OEC-EVs) have been shown to enhance fertilization, protect against polyspermy and sperm entry, and aid embryonic development (11,12). EVs are nano-sized membranous vesicles containing molecular cargo, including micro RNAs (miRNAs), mRNAs, proteins, lipids, and metabolites, that can be easily delivered and fused with cell plasma membranes (13,14). They serve a crucial role in transmitting information and organelles between living cells (15,16) and can modify the epigenetic signature by transferring small molecule RNAs between donor cells (17,18). EVs are present in the female genital tract fluids and are crucial for gamete fertilization and early preimplantation embryonic development (1923). Furthermore, OEC-EVs reduce apoptosis and improve embryo quality by reducing reactive oxygen species (ROS) (24). The first moments of embryonic-maternal communication occur in the oviduct, where both maternal and embryonic EVs are exchanged to prepare for the maternal recognition of pregnancy (25).

Recently, the beneficial effects of OEC-EVs on porcine embryos generated through parthenogenesis and cloning technology (24) and the improved derivation of trophoblast stem cells were reported (26). Thus, the aim of the present study was to mimic the in vivo conditions and reduce polyspermy by investigating the effects of porcine OEC-EVs on the interaction between sperm and oocyte during IVF and subsequent in vitro embryonic development. The potential effects of OEC-EVs on the developmental competence of fertilized oocytes was explored by examining the uptake of EVs by both sperm and oocytes, and the effects on oocyte mitochondrial activity and cortical granule's reaction.

Materials and methods

Chemicals

All chemicals and reagents used in this work were purchased from MilliporeSigma unless otherwise specified.

Oocyte collection and oocyte in vitro maturation (IVM)

Oocyte IVM was performed according to a previously reported method (27). Briefly, porcine ovaries and uteri including the oviducts were obtained from a slaughterhouse in Daejeon City and transported to the laboratory in normal saline solution containing 75 mg/ml penicillin and 50 mg/ml streptomycin. The temperature of the solution was maintained between 34–36°C. Cumulus-oocyte complexes (COCs) were taken from antral follicles (3–8 mm in diameter) with an 18-gauge needle attached to a 10 ml syringe. COCs with three or more layers of cumulus cells and homogeneous ooplasm were selected and washed three times in HEPES-buffered Tyrode's media containing 0.05% (w/v) polyvinyl alcohol (PVA). COCs (n=50) were in vitro matured in 500 µl of maturation medium consisting of bicarbonate-buffered tissue culture medium 199 (TCM-199; Gibco, Thermo Fisher Scientific, Inc.), 10% (v/v) porcine follicular fluid, 0.91 mM sodium pyruvate, 0.57 mM L-cysteine, 10 ng/ml epidermal growth factor and 1 µg/ml insulin with 10 IU/ml equine chorionic gonadotropin (eCG), 10 IU/ml human chorionic gonadotropin (hCG) and 75 µg/ml kanamycin in 4-well dishes (SPL Life Sciences). COCs were incubated at 38.5°C in 5% CO2 in humidified air incubator (Astec Co., Ltd.) for 22 h and then were cultured for an additional 22 h in a hormone-free IVM medium (24).

Porcine oviductal epithelial cell (pOEC) collection

The post-ovulatory oviducts of multiparous female pigs (sows) were obtained from a local butcher and transported to the laboratory according to the aforementioned method. pOECs were isolated by mechanically scraping the oviduct with a slide and then centrifuged three times at 700 × g for 5 min at room temperature in 10 ml of DMEM supplemented with 10% (v/v) FBS (Gibco; Thermo Fisher Scientific, Inc.) and 1% (v/v) antibiotic-antimycotic solution (Gibco; Thermo Fisher Scientific, Inc.). pOECs were cultivated in a 100 mm Falcon tissue culture plate with a culture medium at 39°C and 5% CO2 in a humidified environment. On day 3 of the initial culture, the pOEC outgrowths were examined, after which they were grown for a further 7 days. pOECs were trypsinized in 0.05% trypsin-EDTA, washed in phosphate-buffered saline (PBS) thrice and grown in DMEM without FBS for 48 h to obtain the conditioned medium for isolating OEC-E (24).

EV isolation, purification, and characterization

The supernatant was used to separate EVs using the Total EV Isolation kit after being centrifuged at 2,000 × g for 30 min at room temperature to remove cells and cell debris from the conditioned medium (Invitrogen; Thermo Fisher Scientific, Inc.). The kit reagent was combined with the supernatant, stirred vigorously, and incubated overnight at 2–8°C. The EV pellets were then frozen at −80°C until future investigations. The EV pellet was resuspended in modified Tris-buffered medium (mTBM) for experimental purposes, and the final protein concentration was adjusted to 50 ng/ml using a NanoDrop 2000 (Thermo Scientific; Thermo Fisher Scientific, Inc.). The EVs were visualized using transmission electron microscopy (TEM) after negative staining with 2% uranyl acetate as described previously (24).

ZetaView nanoparticle tracking analysis (NTA)

A ZetaView PMX 110 (Particle Metrix GmbH) instrument was used for NTA as previously described (28). Briefly, 1 ml of diluted EVs pellet (in 1X PBS) was loaded into the device to measure each sample at 11 different positions and two reading cycles per position. After an automated analysis and outlier removal, the mean, median, and mode sizes (indicated as diameter) and the concentrations were calculated using ZetaView SP7 software (version 8.05.14; Particle Metrix) and Microsoft Excel 365 (version 2205, Microsoft Corporation). Device calibration was performed using 100 nm polystyrene particles (Thermo Fisher Scientific, Inc.).

IVF and experimental groups

In vitro matured oocytes with the extruded first polar body were washed twice with mTBM, before being cultured in fresh droplets of mTBM (15 oocytes/50 µl) at 39°C in a humidified atmosphere of 5% CO2. In the OEC EVs group, mTBM contained EVs protein of 50 ng/ml (EVs-mTBM), while the control group was EVs-free. Chilled pig semen was commercially obtained from Darby Genetics Inc., South Korea. Oocytes were co-incubated with 5.0×105 sperm/ml for 20 min. After co-incubation, the attached sperm were discarded from the zona pellucida (ZP) by gentle micro pipetting and the oocytes were washed twice in mTBM. Oocytes were then incubated in mTBM or EVs-mTBM without sperm for an additional 6 h in the same culture conditions. IVF conditions with either OEC-EVs-supplemented or plain culture mediums were compared for polyspermy, cortical granules' reaction, and mitochondrial staining. In vitro embryonic development was monitored after culturing the IVF oocytes in PZM-5 medium (Functional Peptides Research Institute Co. Ltd.) for 7 days at 38.5°C in 5% CO2 and 5% O2 in a humidified incubator.

Penetration and polyspermy

After 18–20 h of IVF, the embryos were fixed using 3.7% (w/v) paraformaldehyde (PFA) for 30 min and washed in PBS/PVA three times at room temperature. The embryos were stained with 10 µg/ml Hoechst 33342 in PBS/PVA for 30 min at room temperature. After staining, the embryos were washed three times in PBS/PVA. The embryos were examined for penetration and pronucleus (PN) formation using a fluorescence microscope. Embryos with one nucleus and polar body (Fig. 1A) were considered non-penetrated. Embryos with one female PN and one male PN in the cytoplasm were considered to be monospermic (Fig. 1B). Embryos with more than one nuclear staining (i.e., sperm or male PNs) were considered to be polyspermic (Fig. 1C).

EV labeling and uptake

After OEC-EV isolation, the OEC EVs were mixed with the lipophilic PKH67 dye according to the manufacturer's instructions, and OEC-EVs were isolated to remove the excess free PKH67 dye according to the manufacturer's protocol (29,30). EVs were then supplemented with sperm or oocytes for 6 h to monitor their uptake using a confocal microscope. For negative control staining, the plain conditioned medium was mixed with PKH67 and processed through the same EV labeling procedure.

Cortical granule (CG) staining

Three times, in vitro matured oocytes, were washed with PBS containing 0.1% (PBS/PVA). The OEC EVs group oocytes were treated for 20 min with OEC EVs in mTBM and the control group was kept in mTBM without EVs. The oocytes were then fixed using 3.7% (w/v) PFA at room temperature for 30 min, followed by three 10 min washes in PBS/PVA per oocyte. This was followed by a 1% Triton X-100 treatment for 30 min and three 10-min washes in PBS/PVA. Oocytes were then cultured for 30 min at room temperature in FITC-labeled Peanut Agglutinin (PNA) diluted 1:500 in PBS in a dark box. Following staining, the oocytes were washed for 10 min in PBS/PVA three times. Finally, the nuclei were stained with DAPI for 10 min, mounted on slides, and images were acquired using a DMi8 confocal microscope (Leica Microsystems GmbH).

Active mitochondria staining

After 18–20 h, the IVF embryos were washed with PBS/PVA three times and were then cultivated for 30 min at 37°C in 500 nM MitoTracker Red CMXRos (cat. no. M7512; Invitrogen; Thermo Fisher Scientific, Inc.) in a dark box. After staining, the embryo was washed three times for 10 min in PBS/PVA. Each embryo was fixed in 3.7% PFA at room temperature for 30 min, followed by three 10 min washes in PBS/PVA. Finally, the nuclei were stained using DAPI for 10 min at room temperature, mounted on slides, and images were acquired using a DMi8 confocal microscope (Leica Microsystems GmbH).

Statistical analysis

Lieven's test and Kolmogorov-Smirnov test were used to confirm the homogeneity of variance and the normality of distribution, respectively. At least 10–20 replicates were performed per experiment. For data normalization, the mean expression level value was calculated from the control group, and the means were normalized to an arbitrary unit (1-fold) by dividing the values against the control group. The image intensities were analyzed using ImageJ (version 1.53k; National Institutes of Health). Data were analyzed using unpaired Student's t-test using SPSS software (SPSS 22.0; IBM Corp). P<0.05 was considered to indicate a statistically significant difference.

Results

Porcine OEC culture and EV characterization

OEC confluent cells were achieved as reported previously (24) and the expression of oviduct epithelial cells' specific gene expression [oviduct-specific glycoprotein (OVGP1)] and proteomics were performed for further confirmation. OEC-EVs were isolated, visualized, and characterized using TEM and NTA, the mean diameter of the isolated EVs was 146.2±57.2 nm, and the mean concentration was 5.3×109 particles/ml (Fig. 1A-C).

Effects of OEC-EVs on sperm penetration and polyspermy

Sperm penetration into the oocyte was detected 18–20 h after IVF. Overall, non-penetration and non-fertilized oocytes (Fig. 2A), penetrated and monospermic fertilized embryos (Fig. 2B), and penetrated and polyspermic abnormally fertilized embryos (Fig. 2C) were observed. The penetration rate did not differ between the OEC-EV and control groups (74.4±1.5 vs. 74.2±2.3, P=0.27). However, the polyspermy rate was significantly lower in the OEC-EV group when compared with the control group (32.9±2.5 vs. 43.8±3.1; P=0.0026; Table I).

Table I.

Effect of OEC-EVs during penetration and polyspermy after in vitro fertilization.

Table I.

Effect of OEC-EVs during penetration and polyspermy after in vitro fertilization.

GroupTotal numberPenetration (% ± SD)Polyspermy (% ± SD)
Control203151 (74.4±4.2)90 (43.8±8.7)
OEC-EVs209155 (74.2±6.6)67 (32.9±7.0)a

a P<0.05 (Control vs. OEC-EVs; unpaired Student's t-test). OEC-EV, oviduct epithelial cell-derived extracellular vesicles; SD, standard deviation. A total of eight replicates were performed (24–27 oocytes each replicate).

Effects of OEC-EVs on IVF embryo development

The cleavage rate differed significantly between the OEC-EV and the control groups (67.6±2.5 vs. 57.3±1.9; P=0.0028). Furthermore, the OEC-EV group had significantly more embryos than the control group (16.4±1.2 vs. 10.2±0.8; P=0.0001; Table II).

Table II.

Effect of OEC-EVs on the development of the in vitro fertilized embryo.

Table II.

Effect of OEC-EVs on the development of the in vitro fertilized embryo.

GroupTotal numberCleaved (% ± SD)Developed to Bl (% ± SD)
Control293168 (57.3±6.2)30 (10.2±2.8)
OEC-EVs318215 (67.6±8.4)a52 (16.4±4.0)a

a P<0.05 (Control vs. OEC-EVs; unpaired Student's t-test). Bl, blastocyst; OEC-EV, oviduct epithelial cell-derived extracellular vesicles; SD, standard deviation. A total of 12 replicates were performed (24–27 oocytes each replicate).

Effects of OEC-EVs on sperm and oocyte communication

In the OEC-EV group, it was observed that OEC-EVs attached to the heads and tails of sperm (Fig. 3) and OEC-EVs that entered the oocyte through the cell membrane (Fig. 4), which was not observed in the control group.

Effects of OEC-EVs on CGs

The CG distribution in the control and OEC-EV groups after FITC-PNA staining were presented (Fig. 5A). The OEC-EV group had significantly higher fluorescence intensity values than those in the control group (3.56±0.47 vs. 2.15±0.24; P<0.05; Fig. 5B).

Effects of OEC-EVs on mitochondrial activity

Mitochondrial activity was detected in both control and OEC-EV-supplemented groups using MitoTracker Red staining (Fig. 6A). The fluorescence values of active mitochondria were higher in the OEC-EV group compared with those in the control group (8.14±0.34 vs. 5.96±0.38; P=0.00007; Fig. 6B).

Discussion

The results of the present study indicated that OEC-EVs support successful porcine IVF by enhancing oocyte mitochondrial activity, reducing polyspermy, and supporting the CG reaction. Polyspermic oocyte penetration is a common reason for decreased IVF success and low blastocyst rates in porcine species (4,9,31). Additionally, previous studies have reported that oxidative stress can alter CG distribution (3234).

First, the effects of EVs on polyspermy were investigated. Amongst other things, oocytes prevent polyspermy by producing CGs that contain enzymes to harden the ZP and prevent subsequent spermatozoa fusion. Recently, EVs have emerged as an important component of several biological fluids, serving a crucial role in communication among different reproductive cells (35). The present study demonstrated that OEC-EVs significantly increased CG content (Fig. 5), which supported previous findings (12,3638). Coy et al (38) and Batista et al (37) reported that incubation of oviductal fluid with porcine IVM oocytes caused ZP hardening, increased ZP resistance to proteolytic digestion, and reduced sperm binding to ZP, subsequently decreasing the polyspermy incidence. The protein cargo contents of OEC-EVs, specifically OVGP1 (22), could be related to increasing monospermy by masking the protease-cleaving sites of ZP proteins and causing ZP hardening (3941). Furthermore, the molecular mechanisms regarding CG formation and release are not well-known. Nonetheless, six genes have been associated with CG distribution after IVM: Rho/Rac guanine nucleotide exchange factor 2, microtubule-associated protein 1B, C-X-C motif chemokine ligand 12, fibronectin 1, DAB adaptor protein 2, and SRY-box transcription factor 9. However, the expression of these genes decreased after IVM (42). The main component of CG is metalloproteinase ovastacin (43), and it is hypothesized that the OEC-EV contents indirectly enhance ovastacin mRNA expression. Importantly, evidence suggests that EVs penetrate the ZP and release their cargo contents inside the ooplasm (36,44), similar to what was observed in the present study (Fig. 4).

The effects of OEC-EVs on oocyte mitochondrial activity were investigated to identify possible mechanisms related to EVs and to improve porcine IVF success. The results of the present study demonstrated that EV treatment increased mitochondrial activity. A previous study suggested that fertilization levels are related to oocyte quality, particularly mitochondrial activity (45). The number and distribution of active mitochondria in an oocyte serve a crucial role in regulating the fertilization of sperm by providing ATP and Ca2+ ion responses (46). Abnormal changes in mitochondria, lipid droplets, calcium release after electro-activation, and the adenosine triphosphate (ATP) and glutathione content in oocytes during aging may result in poor developmental competence of parthenotes (47). OEC-EVs enhanced mitochondrial activity (Fig. 6), providing a better ATP environment for nuclear reprogramming and good conditions for chromosome expansion into chromatin (48,49). Therefore, EVs may initiate several activities in the cytoplasm, which allow the egg to acquire energy and reach a mature state. In vivo and in vitro, oviduct EVs have large amounts of protein related to ATP, adenyl ribonucleotide, purine nucleotide binding, glucose and hexose catabolic processes, and glycolysis (22). Thus, it was hypothesized that OEC-EVs increase mitochondrial activity (16,50) and help provide the large quantity of ATP required for successful fertilization.

Furthermore, the results of the present study indicated that OEC-EVs mediate sperm-oocyte interaction within an in vivo environment. Fallopian tube-secreted EVs contribute to improving the developmental competence and the fertilization of oocytes by reducing the ROS levels, eliciting anti-apoptotic effects, improving lipid metabolism, and increasing the expression of reprogramming-related genes (11,24). Many studies have presented isolation, characterization, and microRNA analyses of oviduct EVs in various species (12,25,35,36,51). Oviduct EV characterization identified proteins with critical roles in the gamete/embryo-oviduct interactions, such as OVGP1, heat shock protein (HSP) 90, HSPA8, HSP70, gelsolin and ezrin (22). In addition, spermatozoa undergo several modifications in the female reproductive tract for the capacitation process to acquire a complete fertilizing ability (52,53). Porcine oviduct EVs attach to sperm membranes (54) and participate in the maintenance of sperm viability and reducing motility, functions associated with the oviduct sperm reservoir (36,55). Oviduct EVs also regulate plasma membrane Ca2+-ATPase 1 to promote sperm motility (7). Moreover, proteins and miRNAs in OEC-EVs may enhance cross-talk in oocyte fertilization (53,56). Ferraz et al (57) reported that oviductal EV supplementation to fresh epididymal spermatozoa enhanced sperm motility and functions. Furthermore, Al-Dossary et al (58) demonstrated that oviductal EVs primarily enhance sperm motility, viability, capacitation, and acrosome reactions during oviduct transit. Therefore, it is hypothesized that OEC-EVs affect sperm penetration and capacitation via the molecular cargo contents, such as OVGP1, myosin heavy chain 9, valosin-containing protein, and annexin A5, or at least in part. Previous studies have observed interactions with these proteins, which are responsible for regulating sperm functions, membrane scaffold/trafficking processes, fertilization, and acrosome reaction (12,22,38,40).

In conclusion, it can be hypothesized that the primary reason for the lower quality of IVF embryos compared to in vivo embryos is due to differences between synthetic medium and pure natural biological secretions. Furthermore, it is suspected that bioactive EVs are a crucial missing link in artificial synthesis. Therefore, it is important to maintain a state as close to that found in vivo as possible to increase the success rate of embryos in vitro. Results of the present study support these hypotheses, as they demonstrated that the addition of OEC-EVs improved porcine IVF by enhancing CG distribution, increasing mitochondrial activity and reducing polyspermy.

Acknowledgements

The authors would like to thank Mr Steve Park at With Instrument Company (Seoul, Republic of Korea) for performing the ZetaView NTA analysis.

Funding

This work was supported by the Ministry of Science, Information, and Communication Technology (MSIT) through the National Research Foundation of Korea (NRF) (grant nos. 2021R1A2C2009294, 2022R1I1A1A01065412) and the Brain Pool program (grant no. 2021H1D3A2A02040098).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XF conceived the study. XF and JC designed the study. XF, BMT, SB, CS, GS, DZ, IMS, SL, XSC, and JC performed the experiments. XF and IMS performed data analysis. XF, IMS, SL, and JC wrote the manuscript. XF and JC confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethical approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Brevini TAL, Antonini S, Cillo F, Crestan M and Gandolfi F: Porcine embryonic stem cells: Facts, challenges and hopes. Theriogenology. 68 (Suppl 1):S206–S213. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Martinez CA, Cambra JM, Maside C, Cuello C, Roca J, Martinez EA, Parrilla I and Gil MA: High pre-freezing sperm dilution improves monospermy without affecting the penetration rate in porcine IVF. Theriogenology. 131:162–168. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Tanihara F, Hirata M, Nguyen NT, Le QA, Hirano T and Otoi T: Effects of concentration of CRISPR/Cas9 components on genetic mosaicism in cytoplasmic microinjected porcine embryos. J Reprod Dev. 65:209–214. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Gil MA, Cuello C, Parrilla I, Vazquez JM, Roca J and Martinez EA: Advances in swine in vitro embryo production technologies. Reprod Domest Anim. 45 (Suppl 2):S40–S48. 2010. View Article : Google Scholar

5 

Grupen CG: The evolution of porcine embryo in vitro production. Theriogenology. 81:24–37. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Nguyen HT, Dang-Nguyen TQ, Somfai T, Men NT, Viet Linh N, Xuan Nguyen B, Noguchi J, Kaneko H and Kikuchi K: Selection based on morphological features of porcine embryos produced by in vitro fertilization: Timing of early cleavages and the effect of polyspermy. Anim Sci J. 91:e134012020. View Article : Google Scholar : PubMed/NCBI

7 

Nguyen HT, Dang-Nguyen TQ, Somfai T, Men NT, Beck-Woerner B, Viet Linh N, Xuan Nguyen B, Noguchi J, Kaneko H and Kikuchi K: Excess polyspermy reduces the ability of porcine oocytes to promote male pronuclear formation after in vitro fertilization. Anim Sci J. 92:e136502021. View Article : Google Scholar : PubMed/NCBI

8 

Romar R, Cánovas S, Matás C, Gadea J and Coy P: Pig in vitro fertilization: Where are we and where do we go? Theriogenology. 137:113–121. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Romero-Aguirregomezcorta J, Soriano-Úbeda C and Matás C: Involvement of nitric oxide during in vitro oocyte maturation, sperm capacitation and in vitro fertilization in pig. Res Vet Sci. 134:150–158. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Li S and Winuthayanon W: Oviduct: Roles in fertilization and early embryo development. J Endocrinol. 232:R1–R26. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Harris EA, Stephens KK and Winuthayanon W: Extracellular vesicles and the oviduct function. Int J Mol Sci. 21:82802020. View Article : Google Scholar : PubMed/NCBI

12 

Alcântara-Neto AS, Fernandez-Rufete M, Corbin E, Tsikis G, Uzbekov R, Garanina AS, Coy P, Almiñana C and Mermillod P: Oviduct fluid extracellular vesicles regulate polyspermy during porcine in vitro fertilisation. Reprod Fertil Dev. 32:409–418. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Kim KM, Abdelmohsen K, Mustapic M, Kapogiannis D and Gorospe M: RNA in extracellular vesicles. Wiley Interdiscip Rev RNA. 8:10.1002/wrna.1413. 2017. View Article : Google Scholar

14 

Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, Liebler DC, Ping J, Liu Q, Evans R, et al: Reassessment of exosome composition. Cell. 177:428–445.e18. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Mathieu M, Martin-Jaular L, Lavieu G and Théry C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 21:9–17. 2019. View Article : Google Scholar : PubMed/NCBI

16 

D'Souza A, Burch A, Dave KM, Sreeram A, Reynolds MJ, Dobbins DX, Kamte YS, Zhao W, Sabatelle C, Joy GM, et al: Microvesicles transfer mitochondria and increase mitochondrial function in brain endothelial cells. J Control Release. 338:505–526. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Willis GR, Reis M, Gheinani AH, Fernandez-Gonzalez A, Taglauer ES, Yeung V, Liu X, Ericsson M, Haas E, Mitsialis SA and Kourembanas S: Extracellular vesicles protect the neonatal lung from hyperoxic injury through the epigenetic and transcriptomic reprogramming of myeloid cells. Am J Respir Crit Care Med. 204:1418–1432. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Mannavola F, D'Oronzo S, Cives M, Stucci LS, Ranieri G, Silvestris F and Tucci M: Extracellular vesicles and epigenetic modifications are hallmarks of melanoma progression. Int J Mol Sci. 21:522019. View Article : Google Scholar : PubMed/NCBI

19 

Bridi A, Perecin F and Silveira JCD: Extracellular vesicles mediated early embryo-maternal interactions. Int J Mol Sci. 21:11632020. View Article : Google Scholar : PubMed/NCBI

20 

Almiñana C and Bauersachs S: Extracellular vesicles in the oviduct: Progress, challenges and implications for the reproductive success. Bioengineering (Basel). 6:322019. View Article : Google Scholar : PubMed/NCBI

21 

Almiñana C and Bauersachs S: Extracellular vesicles: Multi-signal messengers in the gametes/embryo-oviduct cross-talk. Theriogenology. 150:59–69. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Almiñana C, Corbin E, Tsikis G, Alcântara-Neto AS, Labas V, Reynaud K, Galio L, Uzbekov R, Garanina AS, Druart X and Mermillod P: Oviduct extracellular vesicles protein content and their role during oviduct-embryo cross-talk. Reproduction. 154:153–168. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Saadeldin IM, Oh HJ and Lee BC: Embryonic-maternal cross-talk via exosomes: Potential implications. Stem Cells Cloning. 8:103–107. 2015.PubMed/NCBI

24 

Fang X, Tanga BM, Bang S, Seong G, Saadeldin IM, Lee S and Cho J: Oviduct epithelial cells-derived extracellular vesicles improve preimplantation developmental competence of in vitro produced porcine parthenogenetic and cloned embryos. Mol Reprod Dev. 89:54–65. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Mazzarella R, Bastos NM, Bridi A, Del Collado M, Andrade GM, Pinzon J, Prado CM, Silva LA, Meirelles FV, Pugliesi G, et al: Changes in oviductal cells and small extracellular vesicles mirnas in pregnant cows. Front Vet Sci. 8:6397522021. View Article : Google Scholar : PubMed/NCBI

26 

Fang X, Tanga BM, Bang S, Seo C, Kim H, Saadeldin IM, Lee S and Cho J: Oviduct epithelial cell-derived extracellular vesicles improve porcine trophoblast outgrowth. Vet Sci. 9:6092022. View Article : Google Scholar : PubMed/NCBI

27 

Roy PK, Qamar AY, Tanga BM, Bang S, Seong G, Fang X, Kim G, Edirisinghe SL, De Zoysa M, Kang DH, et al: Modified spirulina maxima pectin nanoparticles improve the developmental competence of in vitro matured porcine oocytes. Animals (Basel). 11:24832021. View Article : Google Scholar : PubMed/NCBI

28 

Mehdiani A, Maier A, Pinto A, Barth M, Akhyari P and Lichtenberg A: An innovative method for exosome quantification and size measurement. J Vis Exp. 509742015.PubMed/NCBI

29 

Simonsen JB: Pitfalls associated with lipophilic fluorophore staining of extracellular vesicles for uptake studies. J Extracell Vesicles. 8:15822372019. View Article : Google Scholar : PubMed/NCBI

30 

Takov K, Yellon DM and Davidson SM: Confounding factors in vesicle uptake studies using fluorescent lipophilic membrane dyes. J Extracell Vesicles. 6:13887312017. View Article : Google Scholar : PubMed/NCBI

31 

Wheeler MB, Clark SG and Beebe DJ: Developments in in vitro technologies for swine embryo production. Reprod Fertil Dev. 16:15–25. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Kim JW, Park HJ, Yang SG and Koo DB: Anti-oxidative effects of exogenous ganglioside GD1a and GT1b on embryonic developmental competence in pigs. J Anim Reprod Biotechnol. 35:347–356. 2020. View Article : Google Scholar

33 

Park SH, Jeong PS, Joo YE, Kang HG, Kim MJ, Lee S, Song BS, Kim SU, Cho SK and Sim BW: Luteolin orchestrates porcine oocyte meiotic progression by maintaining organelle dynamics under oxidative stress. Front Cell Dev Biol. 9:6898262021. View Article : Google Scholar : PubMed/NCBI

34 

Jiao X, Ding Z, Meng F, Zhang X, Wang Y, Chen F, Duan Z, Wu D, Zhang S, Miao Y and Huo L: The toxic effects of Fluorene-9-bisphenol on porcine oocyte in vitro maturation. Environ Toxicol. 35:152–158. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Saadeldin IM, Gad A and Mermillod P: Editorial: Biofluid extracellular vesicles and their involvement in animal reproductive physiology. Front Vet Sci. 8:7471382021. View Article : Google Scholar : PubMed/NCBI

36 

Alcântara-Neto AS, Schmaltz L, Caldas E, Blache MC, Mermillod P and Almiñana C: Porcine oviductal extracellular vesicles interact with gametes and regulate sperm motility and survival. Theriogenology. 155:240–255. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Batista RITP, Moro LN, Corbin E, Alminana C, Souza-Fabjan JMG, de Figueirêdo Freitas VJ and Mermillod P: Combination of oviduct fluid and heparin to improve monospermic zygotes production during porcine in vitro fertilization. Theriogenology. 86:495–502. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Coy P, Lloyd R, Romar R, Satake N, Matas C, Gadea J and Holt WV: Effects of porcine pre-ovulatory oviductal fluid on boar sperm function. Theriogenology. 74:632–642. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Coy P and Avilés M: What controls polyspermy in mammals, the oviduct or the oocyte? Biol Rev Camb Philos Soc. 85:593–605. 2010.PubMed/NCBI

40 

Coy P, Cánovas S, Mondéjar I, Saavedra MD, Romar R, Grullón L, Matás C and Avilés M: Oviduct-specific glycoprotein and heparin modulate sperm-zona pellucida interaction during fertilization and contribute to the control of polyspermy. Proc Natl Acad Sci USA. 105:15809–15814. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Avilés M, Coy P and Rizos D: The oviduct: A key organ for the success of early reproductive events. Anim Front. 5:25–31. 2015. View Article : Google Scholar

42 

Kulus M, Kranc W, Jeseta M, Sujka-Kordowska P, Konwerska A, Ciesiółka S, Celichowski P, Moncrieff L, Kocherova I, Józkowiak M, et al: Cortical granule distribution and expression pattern of genes regulating cellular component size, morphogenesis, and potential to differentiation are related to oocyte developmental competence and maturational capacity in vivo and in vitro. Genes (Basel). 11:8152020. View Article : Google Scholar : PubMed/NCBI

43 

Körschgen H, Kuske M, Karmilin K, Yiallouros I, Balbach M, Floehr J, Wachten D, Jahnen-Dechent W and Stöcker W: Intracellular activation of ovastacin mediates pre-fertilization hardening of the zona pellucida. Mol Hum Reprod. 23:607–616. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Saadeldin IM, Kim SJ, Choi YB and Lee BC: Improvement of cloned embryos development by co-culturing with parthenotes: A possible role of exosomes/microvesicles for embryos paracrine communication. Cell Reprogram. 16:223–234. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Babayev E and Seli E: Oocyte mitochondrial function and reproduction. Curr Opin Obstet Gynecol. 27:175–181. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Niu YJ, Zhou W, Nie ZW, Shin KT and Cui XS: Melatonin enhances mitochondrial biogenesis and protects against rotenone-induced mitochondrial deficiency in early porcine embryos. J Pineal Res. 68:e126272020. View Article : Google Scholar : PubMed/NCBI

47 

Hao ZD, Liu S, Wu Y, Wan PC, Cui MS, Chen H and Zeng SM: Abnormal changes in mitochondria, lipid droplets, ATP and glutathione content, and Ca(2+) release after electro-activation contribute to poor developmental competence of porcine oocyte during in vitro ageing. Reprod Fertil Dev. 21:323–332. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Clapier CR, Iwasa J, Cairns BR and Peterson CL: Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat Rev Mol Cell Biol. 18:407–422. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Singhal N, Graumann J, Wu G, Araúzo-Bravo MJ, Han DW, Greber B, Gentile L, Mann M and Schöler HR: Chromatin-remodeling components of the BAF complex facilitate reprogramming. Cell. 141:943–955. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Russell AE, Jun S, Sarkar S, Geldenhuys WJ, Lewis SE, Rellick SL and Simpkins JW: Extracellular vesicles secreted in response to cytokine exposure increase mitochondrial oxygen consumption in recipient cells. Front Cell Neurosci. 13:512019. View Article : Google Scholar : PubMed/NCBI

51 

Machtinger R, Laurent LC and Baccarelli AA: Extracellular vesicles: Roles in gamete maturation, fertilization and embryo implantation. Hum Reprod Update. 22:182–193. 2016.PubMed/NCBI

52 

Qamar AY, Fang X, Bang S, Mahiddine FY, Kim MJ and Cho J: The interplay between exosomes and spermatozoa. Alzahrani FA and Saadeldin IM: Role of Exosomes in Biological Communication Systems. Springer; Singapore: pp. 115–139. 2021

53 

Saadeldin IM: Extracellular vesicles mediate the embryonic-maternal paracrine communication. Alzahrani FA and Saadeldin IM: Role of Exosomes in Biological Communication Systems. Springer; Singapore: pp. 77–97. 2021, View Article : Google Scholar

54 

Du J, Shen J, Wang Y, Pan C, Pang W, Diao H and Dong W: Boar seminal plasma exosomes maintain sperm function by infiltrating into the sperm membrane. Oncotarget. 7:58832–58847. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Kumaresan A, Johannisson A, Humblot P and Bergqvist AS: Effect of bovine oviductal fluid on motility, tyrosine phosphorylation, and acrosome reaction in cryopreserved bull spermatozoa. Theriogenology. 124:48–56. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Bathala P, Fereshteh Z, Li K, Al-Dossary AA, Galileo DS and Martin-DeLeon PA: Oviductal extracellular vesicles (oviductosomes, OVS) are conserved in humans: Murine OVS play a pivotal role in sperm capacitation and fertility. Mol Hum Reprsod. 24:143–157. 2018.PubMed/NCBI

57 

Ferraz MAMM, Carothers A, Dahal R, Noonan MJ and Songsasen N: Oviductal extracellular vesicles interact with the spermatozoon's head and mid-piece and improves its motility and fertilizing ability in the domestic cat. Sci Rep. 9:94842019. View Article : Google Scholar : PubMed/NCBI

58 

Al-Dossary AA, Strehler EE and Martin-DeLeon PA: Expression and secretion of plasma membrane Ca2+-ATPase 4a (PMCA4a) during murine estrus: association with oviductal exosomes and uptake in sperm. PLoS One. 8:e801812013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 27 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fang X, Bang S, Tanga BM, Seo C, Zhou D, Seong G, Saadeldin IM, Lee S, Cui X, Cho J, Cho J, et al: Oviduct epithelial cell‑derived extracellular vesicles promote the developmental competence of IVF porcine embryos. Mol Med Rep 27: 122, 2023
APA
Fang, X., Bang, S., Tanga, B.M., Seo, C., Zhou, D., Seong, G. ... Cho, J. (2023). Oviduct epithelial cell‑derived extracellular vesicles promote the developmental competence of IVF porcine embryos. Molecular Medicine Reports, 27, 122. https://doi.org/10.3892/mmr.2023.13009
MLA
Fang, X., Bang, S., Tanga, B. M., Seo, C., Zhou, D., Seong, G., Saadeldin, I. M., Lee, S., Cui, X., Cho, J."Oviduct epithelial cell‑derived extracellular vesicles promote the developmental competence of IVF porcine embryos". Molecular Medicine Reports 27.6 (2023): 122.
Chicago
Fang, X., Bang, S., Tanga, B. M., Seo, C., Zhou, D., Seong, G., Saadeldin, I. M., Lee, S., Cui, X., Cho, J."Oviduct epithelial cell‑derived extracellular vesicles promote the developmental competence of IVF porcine embryos". Molecular Medicine Reports 27, no. 6 (2023): 122. https://doi.org/10.3892/mmr.2023.13009