Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-26T14:23:14.166Z Has data issue: false hasContentIssue false

Supplementation of porcine in vitro maturation medium with FGF2, LIF, and IGF1 enhances cytoplasmic maturation in prepubertal gilts oocytes and improves embryo quality

Published online by Cambridge University Press:  01 September 2022

María Serrano Albal*
Affiliation:
School of Biosciences, University of Kent, Canterbury, UK
Giuseppe Silvestri
Affiliation:
School of Biosciences, University of Kent, Canterbury, UK
Lucas G. Kiazim
Affiliation:
School of Biosciences, University of Kent, Canterbury, UK
Lucy M. Vining
Affiliation:
Biomolecular Research Group, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, UK
Louisa J. Zak
Affiliation:
Topigs Norsvin Research Center, Beuningen, The Netherlands
Grant A. Walling
Affiliation:
Topigs Norsvin Research Center, Beuningen, The Netherlands
Alexandra M. Haigh
Affiliation:
JSR Genetics Ltd, Southburn, Driffield, East Yorkshire, UK
Simon C. Harvey
Affiliation:
Biomolecular Research Group, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, UK
Katie E. Harvey
Affiliation:
Biomolecular Research Group, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, UK
Darren K. Griffin
Affiliation:
School of Biosciences, University of Kent, Canterbury, UK
*
Author for correspondence: María Serrano Albal. School of Biosciences, University of Kent, Canterbury, UK. E-mail: ms2104@kent.ac.uk
Rights & Permissions [Opens in a new window]

Summary

In porcine in vitro production (IVP) systems, the use of oocytes derived from prepubertal gilts, whilst being commercially attractive, remains challenging due to their poor developmental competence following in vitro maturation (IVM). Follicular fluid contains important growth factors and plays a key role during oocyte maturation; therefore, it is a common supplementation for porcine IVM medium. However, follicular fluid contains many poorly characterized components, is batch variable, and its use raises biosecurity concerns. In an effort to design a defined IVM system, growth factors such as cytokines have been previously tested. These include leukaemia inhibitory factor (LIF), fibroblast growth factor 2 (FGF2), and insulin-like growth factor 1 (IGF1), the combination of which is termed ‘FLI’. Here, using abattoir-derived oocytes in a well established porcine IVP system, we compared follicular fluid and FLI supplementation during both IVM and embryo culture to test the hypothesis that FLI can substitute for follicular fluid without compromising oocyte nuclear and cytoplasmic maturation. We demonstrate that in oocytes derived from prepubertal gilts, FLI supplementation enhances oocyte meiotic maturation and has a positive effect on the quality and developmental competence of embryos. Moreover, for the first time, we studied the effects of follicular fluid and FLI combined showing no synergistic effects.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press

Introduction

The developmental potential of porcine oocytes and embryos is affected by several intrinsic and extrinsic factors that can compromise the efficiency of in vitro production (IVP) systems (Hunter, Reference Hunter2000; Weaver et al., Reference Weaver, Kelly, Kind, Gatford, Kennaway, Herde and van Wettere2013; Teplitz et al., Reference Teplitz, Lorenzo, Maruri, Cruzans, Carou and Lombardo2020). One such factor is the oocyte source; oocytes can be collected from adult donors or prepubertal gilts (Marchal et al., Reference Marchal, Feugang, Perreau, Venturi, Terqui and Mermillod2001). From a commercial standpoint, the use of ovaries from prepubertal gilts is highly attractive as it gives the opportunity to shorten generation times, thereby facilitating the introduction of new genetics more quickly (Sherrer et al., Reference Sherrer, Rathbun and Davis2004). This approach nonetheless presents many challenges and limitations that are mainly related to the ability of the oocytes to undergo nuclear and cytoplasmic maturation during in vitro maturation (IVM) (Marchal et al., Reference Marchal, Feugang, Perreau, Venturi, Terqui and Mermillod2001; Pawlak et al., Reference Pawlak, Cieslak, Warzych, Zejden, Szumacher-Strabel, Molinska-Glura and Lechniak2012). Indeed, in recent years, research has focussed on the improvement of culture conditions for prepubertal porcine oocytes with the aim of assisting them in completing nuclear and cytoplasmic maturation more efficiently (Uhm et al., Reference Uhm, Gupta, Yang, Chung, Min and Lee2010; Appeltant et al., Reference Appeltant, Somfai, Maes, Van Soom and Kikuchi2016; Yuan, et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017; Teplitz et al., Reference Teplitz, Lorenzo, Maruri, Cruzans, Carou and Lombardo2020). Observing the position of the cortical granules (CGs) allows for the determination of cytoplasmic maturation. CGs are specialized secretory vesicles, present only in female germ cells that are randomly distributed throughout the cytoplasm of immature oocytes and migrate towards the cortical cytoplasm during meiotic maturation. Upon fertilization, CGs undergo exocytosis to release their contents with the aim to harden the zona pellucida (Burkart et al., Reference Burkart, Xiong, Baibakov, Jiménez-Movilla and Dean2012). Furthermore, some recent studies have reported that CGs function beyond fertilization, having an effect on the regulation of embryonic cleavage and preimplantation development (Liu, Reference Liu2011; Kulus et al., Reference Kulus, Kranc, Jeseta, Sujka-Kordowska, Konwerska, Ciesiółka, Celichowski, Moncrieff, Kocherova, Józkowiak, Kulus, Wieczorkiewicz, Piotrowska-Kempisty, Skowroński, Bukowska, Machatkova, Hanulakova, Mozdziak and Jaśkowski2020).

Follicular fluid plays a key role during oocyte maturation in sows (Tatemoto et al., Reference Tatemoto, Muto, Sunagawa, Shinjo and Nakada2004), and, as such, sow follicular fluid (sFF) is a common supplement used in pig IVM medium (Pawlak et al., Reference Pawlak, Warzych, Cieslak, Malyszka, Maciejewska, Madeja and Lechniak2018). It is known to support in vitro oocyte development by providing the culture environment with a variety of important growth factors (Lédée et al., Reference Lédée, Lombroso, Lombardelli, Selva, Dubanchet, Chaouat, Frankenne, Foidart, Maggi, Romagnani, Ville and Piccinni2008). The growth factors found in sFF are naturally produced by ovarian somatic cells during final follicle development, acting as both autocrine and paracrine mediators of ovarian function and regulating oocyte maturation and oocyte developmental competence (Song et al., Reference Song, Kang, Maeng, Ock, Lee, Yoo, Jeon and Rho2011). However, the complete constitution of sFF is unknown and its use results in a non-defined culture system and raises potential biosecurity concerns. Batch variation has also been demonstrated in sFF and its composition is known to change with the donor’s age and stage of the follicles from which it is recovered (Sun et al., Reference Sun, Ping, Li, Sun, Yi, Chen, Li, Wang and Zhou2011). Furthermore, variation in the concentrations of any of the components of sFF can affect IVM outcomes and therefore oocyte developmental competence (De Oliveira et al., Reference De Oliveira, Lopes and Rodrigues2006; Pawlak et al., Reference Pawlak, Warzych, Cieslak, Malyszka, Maciejewska, Madeja and Lechniak2018).

In an effort to design efficient, but defined IVM culture medium, growth factors such as certain cytokines have been tested, alone or in combination, for the culture of porcine oocytes. In particular, leukaemia inhibitory factor (LIF) and fibroblast growth factor 2 (FGF2) have been shown to facilitate meiotic progress during maturation, promoting oocyte quality and its subsequent ability to achieve fertilization. It has been shown that LIF phosphorylates MAPK3/1 and STAT3 in the oocytes, which are important pathways during in vitro maturation (Dang-Nguyen et al., Reference Dang-Nguyen, Haraguchi, Kikuchi, Somfai, Bodó and Nagai2014; Mo et al., Reference Mo, Wu, Yuan, Jia, Liu, Zhu and Hou2014). FGF2 acts as a cofactor, promoting cumulus cell survival and extracellular matrix quality during IVM (Barros et al., Reference Barros, Lima, Soares, Sanches, Price and Buratini2019; Du et al., Reference Du, Davis, Chen, Li, Cao, Sun, Shao, Lin, Wang, Yang and Hua2021). Insulin-like growth factor 1 (IGF1) also has been found to improve IVM outcomes and increase cell survival in response to stress (Oberlender et al., Reference Oberlender, Murgas, Zangeronimo, da Silva, Menezes, Pontelo and Vieira2013). IGF1 enables the expansion of porcine cumulus cells in response to FSH, promoting the synthesis and retention of hyaluronic acid in porcine COCs, also activated by the MAPK3/1 pathway (Singh and Armstrong, Reference Singh and Armstrong1997; Němcová et al., Reference Němcová, Nagyová, Petlach, Tománek and Procházka2007).

Interestingly, the use of these three cytokines during IVM, known together as FLI (Yuan, et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017), positively affects outcomes by improving oocyte quality, embryonic development, embryo transfer outcomes and, in cattle, embryo cryosurvival (Yuan et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017; Stoecklein et al., Reference Stoecklein, Ortega, Spate, Murphy and Prather2021).

Here, we performed a direct comparison between sFF and FLI supplementation of IVM medium using oocytes from prepubertal gilts and semen stored in extender. We tested the hypothesis that FLI can efficiently substitute for sFF during IVM without compromising oocyte nuclear and/or cytoplasmic maturation. Moreover, for the first time we studied the effects of combining sFF and FLI in IVM of oocytes to test the hypothesis that there is a synergistic effect between these components that would suggest a role for additional growth factors that may be present in sFF but absent in FLI.

Materials and methods

All chemicals and reagents used were purchased from Sigma-Aldrich (Gillingham, UK) except when specified otherwise.

Oocyte collection and IVM

Prepubertal gilt ovaries were collected and transported to the laboratory within 5 h in a sealed bag floated in water at 30–35°C. The animals had an approximate weight of 160 kg at slaughter, and their prepubertal status was confirmed by the absence of developed ovarian corpora lutea. Prior to aspiration, the ovaries were washed two or three times in 1× phosphate-buffered saline (PBS) and kept in a water bath at 28°C. The cumulus–oocyte complexes (COCs) were collected by manual aspiration from non-atretic follicles (3–6 mm) using a non-pyrogenic/non-toxic syringe (Henke-Sass Wolf GmbH, Tuttlingen, Germany) fitted with an 18-gauge needle. The follicular contents were washed three times in a modified HEPES-buffered Porcine X Medium [PXM; Yoshioka et al., Reference Yoshioka, Suzuki and Onishi2008; 108 mM NaCl, 10 mM KCl, 0.35 mM KH2PO4, 0.40 mM MgSO4, 5.0 mM NaHCO3, 25 mM HEPES, 0.2 mM sodium pyruvate, 2.0 mM calcium lactate, 4 mg/ml bovine serum albumin (BSA)]; warmed at 38°C.

Selected COCs were washed three times in porcine oocyte medium [POM; Yoshioka et al., Reference Yoshioka, Suzuki and Onishi2008; 108 mM NaCl, 10 mM KCl, 0.3 5 mM KH2PO4, 0.4 mM MgSO4, 25 mM NaHCO3, 5.0 mM glucose, 0.2 mM sodium pyruvate, 2.0 mM calcium lactate, 2.0 mM glutamine, 5.0 mM hypotaurine, 0.1 mM cysteine, 20 µl/ml Basal Medium Eagle (BME) amino acids 50×, 10 µl/ml minimum essential medium (MEM) non-essential amino acids 100×, 10 ng/ml EGF, 50 µM β-mercaptoethanol, 10 µg/ml gentamycin, 4 mg/ml BSA] previously equilibrated overnight at 38.5°C in a saturated humidity atmosphere of 5.5% CO2 in air. COCs were randomly assigned to groups of 50 for treatment with different supplements: FLI, sFF, both FLI and sFF, or left unsupplemented (please refer to ‘Experimental design’ for details). COCs were cultured for 20 h in POM supplemented with FSH (0.5 IU/ml), LH (0.5 IU/ml) and dbc-AMP (0.1 mM). Subsequently, COCs were cultured in the same medium but without hormones and dbc-AMP for a further 24 h at 38.5°C and 5.5% CO2 in humidified air.

In vitro fertilization (IVF) and culture

Extended boar semen [for commercial artificial insemination (AI)] was supplied by JSR Genetics Ltd (Southburn, UK). Sperm preparation was performed using a 35%/70% BoviPure discontinuous density gradient system (Nidacon, Göthenborg, Sweden) following the manufacturer’s instructions.

Matured oocytes were washed twice in porcine gamete medium (PGM; Yoshioka et al., Reference Yoshioka, Suzuki and Onishi2008; 108 mM NaCl, 10 mM KCl, 0.35 mM KH2PO4, 0.4 mM MgSO4, 25 mM NaHCO3, 5.0 mM glucose, 0.2 mM sodium pyruvate, 2.0 mM calcium lactate, 2.5 mM theophylline, 1 µM adenosine, 0.25 µM l-cysteine, 10 µg/ml gentamycin, 4 mg/ml BSA) and were incubated with sperm for 2 h. Following this co-incubation, oocytes were moved to a clean well in PGM for another 2 h to minimize the risk of polyspermy.

After IVF, presumptive zygotes were denuded and washed twice in porcine zygote medium 5 (PZM5; Yoshioka et al., Reference Yoshioka, Suzuki and Onishi2008; 108 mM NaCl, 1 mM KCl, 0.35 mM KH2PO4, 0.4 mM MgSO4, 25 mM NaHCO3, 0.2 mM sodium pyruvate, 2.0 mM calcium lactate, 2.0 mM glutamine, 5.0 mM hypotaurine, 20 µl/ml BME amino acids 50×, 10 µl/ml MEM non-essential amino acids 100×, 10 µg/ml gentamycin, 4 mg/ml BSA) before being transferred to a final 500 µl well of PZM5 supplemented with FLI. Individual wells were overlaid with mineral oil and plates were incubated for 6 days at 38.5°C, in 5.5% CO2 and 6% O2 in humidified air.

Evaluation of nuclear stage and cortical granule distribution

Oocytes were fixed with 4% paraformaldehyde (PFA) in PBS for 30 min at 4°C, and then washed three times in PBS containing 0.3% BSA and 100 mM glycine for 5 min. After a 5-min treatment with 0.1% Triton X-100 in PBS, oocytes were washed two additional times in PBS (5 min each). To stain the CG, oocytes were cultured in 100 mg/ml fluorescein isothiocyanate-labelled peanut agglutinin (FITC-PNA; Invitrogen™, Inchinnan, UK) in PBS for 30 min in a dark box. After staining, oocytes were washed three times in PBS with 0.3% BSA and 0.01% Triton X-100 in PBS. The oocytes were then stained with 10 mg/ml Hoechst H3570 (Invitrogen™) for 10 min, mounted on non-fluorescent glass slides and visualized using an Olympus BX61 epifluorescence microscope equipped with a cooled charge-coupled device (CCD) camera. Images were taken using 4′,6-diamidino-2-phenylindole (DAPI) and FITC filters at ×200 total magnification using SmartCapture3 software (Digital Scientific UK, Cambridge, UK). COCs were classified as mature when they displayed a dye-stained metaphase plate and a polar body; otherwise, they were classified as immature. The distribution of CGs was categorized in three ways as: central, peripheral or complete (Figure 1), where: central distribution indicated a homogenous distribution of CGs throughout the cytoplasm; peripheral distribution, indicated that CGs had begun to localize subjacent to the oolemma, or complete distribution; and where CGs were concentrated subjacent to the oolemma and around the polar body (an indicator of complete cytoplasmic maturation).

Figure 1. Effect of each treatment on the proportion of mature oocytes. Mean of the effects of the studied supplements on nuclear maturation of prepubertal gilt oocytes after 44 h of culture. Error bars show the 95% confidence interval. Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.

Evaluation of blastocysts

Blastocyst morphological appearance was assessed and scored using three grades: (1) excellent: fully expanded blastocyst, spherical, regular border, symmetrical with uniform size cells, obvious inner cell mass (ICM) and densely populated trophectoderm (TE); (2) good, expanded blastocyst with few small blastomeres, fewer cells forming the ICM/TE; (3) poor: expanded or less developed blastocyst with numerous extruded blastomeres, loosely populated TE and possible ICM. After grading, blastocysts from each treatment group were fixed in 4% PFA for 30 min at 4°C and stained with Hoechst H3570 (Invitrogen™) for cell counts. Blastocysts were visualized under an Olympus BX61 epifluorescence microscope as described in the previous section.

Experimental design

Experiment 1: Oocyte nuclear maturation following supplementation of IVM medium FLI or sFF

This experiment was designed to study the effect of FLI supplementation during the IVM culture period on the meiotic competence of COCs. Four groups of ∼50 immature COCs were treated during IVM. The first group was supplemented with a combination of three cytokines (Novus Biologicals, Oxon, UK), ‘FLI’, prepared as described by Yuan et al. (Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017). The second group was treated with 10% sFF. The third group was treated with the combination of FLI and 10% sFF, to study a possible synergistic effect. The fourth group of COCs was kept unsupplemented and served as a control. The experiment was repeated four times with 40–50 oocytes per treatment in each replicate (N = 763).

Experiment 2: CG distribution (cytoplasmic maturation) following supplementation of IVM medium with FLI or sFF

This experiment aimed to assess CG distribution as an indicator for the level of cytoplasmic maturation of the COCs depending on the treatment during IVM cultured. Treatment groups were as described in Experiment 1. This experiment was replicated three times with 40–50 oocytes per treatment per replicate (N = 465). In this case, a greater proportion of oocytes with ‘complete’ CGs (i.e. neither predominantly peripheral nor central) is the most favourable outcome.

Experiment 3: Post-fertilization outcomes following supplementation of IVM medium with FLI or sFF

The third experiment was designed to test the effects of different supplementations during the IVM process on the oocyte’s developmental competence following IVF. This was evaluated using blastocyst rates (blastocyst per inseminated oocyte), blastocyst morphology, and blastocyst cell counts. To estimate the total cell counts, nuclei were stained using Hoechst H3570. The live image of embryo was sequentially divided into smaller sections that could be individually brought into focus to improve the accuracy of the count. The same four IVM groups described above (FLI, sFF, FLI + sFF, and control) were used. The experiment was repeated seven times with 40–50 presumptive zygotes per group per replicate (N = 1133). To evaluate the embryo quality of each group, 149 blastocysts were assessed.

Statistical analysis

Data were analyzed using SPSS software (Version 26, IBM). The statistical analysis of oocyte nuclear maturation and blastulation rates were completed by fitting a binomial generalized linear model (GLM) with logit link functions. The numbers of blastomeres per blastocyst were log transformed and were assessed using analysis of variance (ANOVA). When analyzing cytoplasmic maturation and the morphology evaluation of the embryos the data were tested using Kruskal–Wallis test. When an interaction was detected amongst the variables, multiple comparisons were completed using the Bonferroni correction. The data in tables are presented as mean ± standard error of the mean (SEM), whilst in the graphs they appear as mean with the error bars showing the 95% confidence interval of each variable. Results were considered to be statistically significant when P-values were < 0.05.

Results

Effect of FLI and/or sFF supplementation on nuclear maturation

The assessment of the nuclear stage of 763 oocytes after maturation (Figure 1) showed the highest proportion of mature oocytes in the group treated with FLI (70.9%). The use of sFF during IVM gave an outcome slightly lower than the group with no supplementation (control), 61.5% and 62.3% respectively. The lowest rate was found in the combination group (sFF + FLI) with a 60.3%. However, due to the intergroup variation, these differences were not statistically significant (GLM; χ1 = 1.059; P > 0.05).

Effect of FLI and/or sFF supplementation on oocyte CG distribution (cytoplasmic maturation)

The analysis of the distribution of the CGs after IVM culture (Table 1, Figure 2) showed a higher incidence of ‘complete’ distribution (Figure 2c) on the groups treated with FLI (48.8%), second on the combination of FLI and sFF (43.0%), third in the control group (32.7%) and finally for sFF (28.7%). The incidence of a peripheral CG distribution (Figure 2b) was similar in the four groups. Intergroup comparison showed that there were significant differences between the groups (Kruskal–Wallis; H3 = 10.995; P = 0.012).

Table 1. Distribution of cortical granules (cytoplasmic maturation) in oocytes after 44 h of in vitro maturation

Data are shown as mean ± standard error of the mean (SEM) (N = 3 replicates for each group).

a,b Different superscript letters indicate significant differences amongst the groups.

* Kruskal–Wallis test (H3 = 10.995).

Figure 2. Cortical granule (CG) distribution in oocytes after IVM. GV: Homogenous distribution of CGs throughout the cytoplasm in a germinal vesicle (GV) stage oocyte denoted with the arrow. MI: Peripheral distribution on metaphase I (MI) stage oocyte, CGs begin to locate subjacent to the oolemma. MII: Complete distribution on a mature oocyte (metaphase II, MII), CGs are concentrated at the periphery of the cytoplasm and around the polar body (PB). CGs stained in green (FITC-PNA), DNA stained in blue (Hoechst), ×200 total magnification. Each column displays an individual oocyte; the top row displays a combined image, with the subsequent images showing individual fluorophores (FITC or Hoechst).

Effect of the use of FLI and/or sFF during IVM on embryo developmental competence

After fertilization, the highest rate of blastocyst formation per inseminated oocyte (Figure 3) was found in the group treated with FLI (15.7%), followed by the sFF treatment group (14.3%). The proportion of blastocysts from the combination group (sFF + FLI) was similar to the control group (12.0% and 10.8%, respectively). The differences on the proportion of blastocyst formed between the groups did not show any significant difference after statistical analysis (GLM; χ1 = 0.044; P >0.05).

Figure 3. Proportion of blastocyst formation after the different treatments on in vitro maturation. Mean of the effects of the studied supplements on blastulation rates after matured prepubertal gilt oocytes were fertilized. Error bars show the 95% confidence interval. Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.

The morphological assessment of the blastocyst (Figure 4) generated by each group revealed that the oocytes treated with FLI during IVM developed into better quality embryos (Table 2). The highest proportion of the embryos with excellent morphology (Figure 4a,d) came from the FLI treatment group (37.0 ± 12.1%) and this was significantly higher than in the other groups (Kruskal–Wallis; H3 = 8.162; P = 0.043). This group also had the lower proportion of poor-quality embryos (19.6 ± 13.4%). In contrast, the group with a higher percentage of poor-quality embryos (Fig. 4c,f) was the control group (34.5 ± 13.6%). Interestingly, the group cultured in a combination of sFF and FLI had the second highest proportion of excellent quality embryos (29.4 ± 4.8%), but also the second highest proportion of poor-quality blastocyst (29.4 ± 6.6%).

Figure 4. Pig blastocyst morphology. (A–C) Images taken by the use of epifluorescence microscopy at ×200 total magnification. Nuclei in blue (Hoechst staining). (D–F) Phase-contrast optical microscopy images at ×200 total magnification. (A, D) Blastocysts of excellent morphology (grade 1); (B, E) blastocysts of good morphology (grade 2); (C, F) blastocysts of poor morphology (grade 3). Arrows denotate the inner cell mass (ICM) of the blastocysts.

Table 2. Blastocyst quality after the different treatments on in vitro maturation

Data are shown as mean ± SEM (N = 7 replicates for each group).

a,b Different superscript letters indicate significant differences amongst the groups.

* Kruskal–Wallis test (H3 = 8.162).

To further assess the developmental competence, the number of cells was counted for each blastocyst. The average number of cells per blastocyst (Figure 5) was highest in the FLI group (62.1 cells per blastocyst), followed by the control (57.1 cells per blastocyst) and the lower number of cells using sFF as treatment (52.7 and 52.2 cells per blastocyst on the sFF and sFF + FLI groups, respectively). Comparison of treatment groups indicated that they did not differ (ANOVA; F3 = 1.039; P > 0.05).

Figure 5. Average number of cells per blastocyst. Mean of the effects of the different combinations on the number of cells per blastocyst after matured prepubertal gilt oocytes were fertilized. Error bars show the 95% confidence interval (N = 73). Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.

Discussion

Whilst FLI supplementation during IVM always resulted in improved outcomes compared with control experiments and often to other treatment groups, not all findings were statistically significant. However, the use of FLI consistently yielded better results. By contrast, the use of sFF alone had very little effect on oocyte maturation, yielding similar results to the control group, and when used in combination with FLI outcomes were only marginally improved.

The areas in which statistically significant differences were observed include cytoplasmic maturation (distribution of CGs) and blastocyst quality, meaning that the positive effect of FLI supplementation also extended to in vitro culture (IVC). However, our results showed that FLI supplementation did not statistically improve nuclear maturation, blastulation rate, or the number of cells per blastocyst (contrary to a previous study by Yuan et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017). The lack of significance could be influenced by statistical power, as the number of blastocysts analyzed differed from the total number of blastocysts obtained; the difference in this number is a result of fragile blastocysts being destroyed during the fixing process.

It is widely accepted that the survival rate of porcine IVP blastocysts is very low because of their poor quality (Zijlstra et al., Reference Zijlstra, Kidson, Schoevers, Daemen, Tharasanit, Kuijk, Hazeleger, Ducro-Steverink, Colenbrander and Roelen2008). Here we show that the use of FLI during IVM improved the synchrony between nuclear and cytoplasmic maturation, improving overall oocyte quality (Marchal et al., Reference Marchal, Feugang, Perreau, Venturi, Terqui and Mermillod2001; Pawlak et al., Reference Pawlak, Cieslak, Warzych, Zejden, Szumacher-Strabel, Molinska-Glura and Lechniak2012). This synchrony could positively influence the zygote’s developmental competence and its ability to undertake normal cell division, resulting in the symmetrical division of the blastomeres and lower fragmentation rates, which is demonstrated in the results. FLI supplementation not only improved the quality of the resulting embryos that developed to blastocyst stage, but has previously been shown to enhance the efficiency of cattle and sheep IVP and embryo quality (Stoecklein et al., Reference Stoecklein, Ortega, Spate, Murphy and Prather2021; Tian et al., Reference Tian, Qi, Yan and Hou2021). The viability to implant and birth rates of the embryos have not been tested in this study. However, other publications have shown an increase in the postimplantation viability of embryos treated with growth factors (Zheng et al., Reference Zheng, Wen, Song, Wang, Chen and Magness2008; Biswas et al., Reference Biswas, So, Hwang, Yoon, Kim, Kim and Hyun2018).

It is common to have supplements in the medium that act additively to improve in vitro embryo production, with publications showing an increase in the blastulation rates after treatment with individual cytokines, separately as well as in combination (Valleh et al., Reference Valleh, Zandi, Rasmussen and Hyttel2017; Yuan, et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017; Stoecklein et al., Reference Stoecklein, Ortega, Spate, Murphy and Prather2021). Furthermore, Liu et al. (Reference Liu, Hao, Li, Zhou, Zhu, Bu, Liu, Hou, Zhang and Miao2020) demonstrated the positive effect that cytokines had on promoting porcine oocyte maturation and Redel et al. (Reference Redel, Spate, Yuan, Murphy, Roberts and Prather2021) showed that the combination of FLI with gonadotropins enhanced oocyte development and improved cumulus cell expansion. Procházka et al. (Reference Procházka, Bartková, Němcová, Murín, Gad, Marcollová, Kinterová, Lucas-Hahn and Laurinčík2021) demonstrated a similar activation pattern of MAPK3/1 between medium supplemented with or without FLI, showing only an increase in MAPK3/1 phosphorylation during the first hour of culture in the FLI medium. This suggests that COCs cultured with FLI experience enhanced MAPK3/1 stimulation that resulted in the development of highly competent oocytes. Our data demonstrate that combining FLI and sFF did not have a synergistic effect on IVP outcomes, which could be attributed to factors such as the variation in sFF composition (De Oliveira et al., Reference De Oliveira, Lopes and Rodrigues2006; Sun et al., Reference Sun, Ping, Li, Sun, Yi, Chen, Li, Wang and Zhou2011). The donor’s origin, age, and the stage of the follicles from the ovary may be one of the reasons why the results shown in this publication differed from the information published previously on the effect of FLI on porcine maturation (Yuan, et al., Reference Yuan, Spate, Redel, Tian, Zhou, Prather and Roberts2017; Redel et al., Reference Redel, Spate, Yuan, Murphy, Roberts and Prather2021).

Porcine IVP has, to the disappointment of many in the industry, lagged behind its bovine counterpart. By common consent, porcine IVP is technically more challenging at all stages than bovine (or even human) and incremental improvements, such as those reported in this study, are always welcome. The potential for routine IVP in the pig breeding industry is considerable in terms of biosecurity, cost and environmental benefits for transport of both male and female genetics (nationally and internationally). Moreover, using peri-pubertal gilts as a source of ovaries, as in this study, has great potential for the rapid introduction of new genetics through the shortening of generation intervals. Finally, as a resource for gene editing and biobanking, porcine embryos hold great potential provided that the barriers to their production can be overcome. The results of this study therefore represent a significant advance. Given the added problems of batch variability and biosecurity associated with the use of sFF, our future strategies will rely on the use of FLI alone.

Acknowledgements

The authors wish to thanks the staff of Cranswick Country Foods (Hull, UK) for their assistance in sourcing the samples required for this study.

Author contributions

Conceptualization: MSA, GS, DKG, LJZ; formal analysis: MSA, GS. funding acquisition: DKG; investigation: MSA, GS, LGK, LMV; methodology: MSA, GS; resources: AMH, LJZ, GAW; supervision: DKG, LJZ, KEH, SCH; writing – original draft: MSA; writing – review and editing: MSA, GS, LGK, LMV, KEH, SCH, DKG, LJZ. All the authors have read and agreed to publish this version of the manuscript.

Funding

This work was supported by the Biotechnology and Biological Sciences Research Council (BBSRC) LINK awards scheme (BB/R00708X/1).

Institutional review board statement

This project involved the use of abattoir-derived material only and so did not fall within the Animals (Scientific Procedures) Act 1986. It therefore did not require review by an Animal Welfare and Ethical Review Body.

Informed consent statement

Not applicable.

Conflicts of interest

The authors declare no conflict of interest.

References

Appeltant, R., Somfai, T., Maes, D., Van Soom, A. and Kikuchi, K. (2016). Porcine oocyte maturation in vitro: Role of cAMP and oocyte-secreted factors—A practical approach. Journal of Reproduction and Development, 62(5), 439449. doi: 10.1262/jrd.2016-016 CrossRefGoogle ScholarPubMed
Barros, R. G., Lima, P. F., Soares, A. C. S., Sanches, L., Price, C. A. and Buratini, J. (2019). Fibroblast growth factor 2 regulates cumulus differentiation under the control of the oocyte. Journal of Assisted Reproduction and Genetics, 36(5), 905913. doi: 10.1007/s10815-019-01436-7 CrossRefGoogle ScholarPubMed
Biswas, D., So, K. H., Hwang, S. U., Yoon, J. D., Kim, M., Kim, D. Y. and Hyun, S. H. (2018). Embryotropic effects of vascular endothelial growth factor on porcine embryos produced by in vitro fertilization. Theriogenology, 120, 147156. doi: 10.1016/j.theriogenology.2018.07.024 CrossRefGoogle ScholarPubMed
Burkart, A. D., Xiong, B., Baibakov, B., Jiménez-Movilla, M. and Dean, J. (2012). Ovastacin, a cortical granule protease, cleaves ZP2 in the zona pellucida to prevent polyspermy. Journal of Cell Biology, 197(1), 3744. doi: 10.1083/jcb.201112094 CrossRefGoogle ScholarPubMed
Dang-Nguyen, T. Q., Haraguchi, S., Kikuchi, K., Somfai, T., Bodó, S. and Nagai, T. (2014). Leukemia inhibitory factor promotes porcine oocyte maturation and is accompanied by activation of signal transducer and activator of transcription 3. Molecular Reproduction and Development, 81(3), 230239. doi: 10.1002/mrd.22289 CrossRefGoogle ScholarPubMed
De Oliveira, A. T., Lopes, R. F. and Rodrigues, J. L. (2006). Gene expression and developmental competence of bovine embryos produced in vitro with different serum concentrations. Reproduction in Domestic Animals, 41(2), 129136. doi: 10.1111/j.1439-0531.2006.00653.x CrossRefGoogle Scholar
Du, C., Davis, J. S., Chen, C., Li, Z., Cao, Y., Sun, H., Shao, B. S., Lin, Y. X., Wang, Y. S., Yang, L. G. and Hua, G. H. (2021). FGF2/FGFR signaling promotes cumulus–oocyte complex maturation in vitro . Reproduction, 161(2), 205214. doi: 10.1530/REP-20-0264 CrossRefGoogle ScholarPubMed
Hunter, M. G. (2000). Oocyte maturation and ovum quality in pigs. Reviews of Reproduction, 5(2), 122130. doi: 10.1530/ror.0.0050122 CrossRefGoogle ScholarPubMed
Kulus, M., Kranc, W., Jeseta, M., Sujka-Kordowska, P., Konwerska, A., Ciesiółka, S., Celichowski, P., Moncrieff, L., Kocherova, I., Józkowiak, M., Kulus, J., Wieczorkiewicz, M., Piotrowska-Kempisty, H., Skowroński, M. T., Bukowska, D., Machatkova, M., Hanulakova, S., Mozdziak, P., Jaśkowski, J. M., et al. (2020). 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, 11(7), 815. doi: 10.3390/genes11070815 CrossRefGoogle ScholarPubMed
Lédée, N., Lombroso, R., Lombardelli, L., Selva, J., Dubanchet, S., Chaouat, G., Frankenne, F., Foidart, J. M., Maggi, E., Romagnani, S., Ville, Y. and Piccinni, M. P. (2008). Cytokines and chemokines in follicular fluids and potential of the corresponding embryo: The role of granulocyte colony-stimulating factor. Human Reproduction, 23(9), 20012009. doi: 10.1093/humrep/den192 CrossRefGoogle ScholarPubMed
Liu, M. (2011). The biology and dynamics of mammalian cortical granules. Reproductive Biology and Endocrinology: RB&E, 9(1), 149. doi: 10.1186/1477-7827-9-149 CrossRefGoogle ScholarPubMed
Liu, X., Hao, Y., Li, Z., Zhou, J., Zhu, H., Bu, G., Liu, Z., Hou, X., Zhang, X. and Miao, Y. L. (2020). Maternal cytokines CXCL12, VEGFA, and WNT5A promote porcine oocyte maturation via MAPK activation and canonical WNT inhibition. Frontiers in Cell and Developmental Biology, 8, 578. doi: 10.3389/fcell.2020.00578 CrossRefGoogle ScholarPubMed
Marchal, R., Feugang, J. M., Perreau, C., Venturi, E., Terqui, M. and Mermillod, P. (2001). Meiotic and developmental competence of prepubertal and adult swine oocytes. Theriogenology, 56(1), 1729. doi: 10.1016/s0093-691x(01)00539-8 CrossRefGoogle ScholarPubMed
Mo, X., Wu, G., Yuan, D., Jia, B., Liu, C., Zhu, S. and Hou, Y. (2014). Leukemia inhibitory factor enhances bovine oocyte maturation and early embryo development. Molecular Reproduction and Development, 81(7), 608618. doi: 10.1002/mrd.22327 CrossRefGoogle ScholarPubMed
Němcová, L., Nagyová, E., Petlach, M., Tománek, M. and Procházka, R. (2007). Molecular mechanisms of insulin-like growth factor 1 promoted synthesis and retention of hyaluronic acid in porcine oocyte-cumulus complexes. Biology of Reproduction, 76(6), 10161024. doi: 10.1095/biolreprod.106.057927 CrossRefGoogle ScholarPubMed
Oberlender, G., Murgas, L. D. S., Zangeronimo, M. G., da Silva, A. C., Menezes, Tde A., Pontelo, T. P. and Vieira, L. A. (2013). Role of insulin-like growth factor-I and follicular fluid from ovarian follicles with different diameters on porcine oocyte maturation and fertilization in vitro . Theriogenology, 80(4), 319327. doi: 10.1016/j.theriogenology.2013.04.018 CrossRefGoogle ScholarPubMed
Pawlak, P., Cieslak, A., Warzych, E., Zejden, Z., Szumacher-Strabel, M., Molinska-Glura, M. and Lechniak, D. (2012). No single way to explain cytoplasmic maturation of oocytes from prepubertal and cyclic gilts. Theriogenology, 78(9), 20202030. doi: 10.1016/j.theriogenology.2012.07.023 CrossRefGoogle ScholarPubMed
Pawlak, P., Warzych, E., Cieslak, A., Malyszka, N., Maciejewska, E., Madeja, Z. E. and Lechniak, D. (2018). The consequences of porcine IVM medium supplementation with follicular fluid become reflected in embryo quality, yield and gene expression patterns. Scientific Reports, 8(1), 15306. doi: 10.1038/s41598-018-33550-4 CrossRefGoogle ScholarPubMed
Procházka, R., Bartková, A., Němcová, L., Murín, M., Gad, A., Marcollová, K., Kinterová, V., Lucas-Hahn, A. and Laurinčík, J. (2021). The role of MAPK3/1 and AKT in the acquisition of high meiotic and developmental competence of porcine oocytes cultured in vitro in FLI medium. International Journal of Molecular Sciences, 22(20), 11148. doi: 10.3390/ijms222011148 CrossRefGoogle ScholarPubMed
Redel, B. K., Spate, L. D., Yuan, Y., Murphy, C. N., Roberts, R. M. and Prather, R. S. (2021). Neither gonadotropin nor cumulus cell expansion is needed for the maturation of competent porcine oocytes in vitro . Biology of Reproduction, 105(2), 533542. doi: 10.1093/biolre/ioab090 CrossRefGoogle Scholar
Sherrer, E. S., Rathbun, T. J. and Davis, D. L. (2004). Fertilization and blastocyst development in oocytes obtained from prepubertal and adult pigs. Journal of Animal Science, 82(1), 102108. doi: 10.2527/2004.821102x CrossRefGoogle ScholarPubMed
Singh, B. and Armstrong, D. T. (1997). Insulin-like growth factor-1, a component of serum that enables porcine cumulus cells to expand in response to follicle-stimulating hormone in vitro . Biology of Reproduction, 56(6), 13701375. doi: 10.1095/biolreprod56.6.1370 CrossRefGoogle ScholarPubMed
Song, H. J., Kang, E. J., Maeng, G. H., Ock, S. A., Lee, S. L., Yoo, J. G., Jeon, B. G. and Rho, G. J. (2011). Influence of epidermal growth factor supplementation during in vitro maturation on nuclear status and gene expression of canine oocytes. Research in Veterinary Science, 91(3), 439445. doi: 10.1016/j.rvsc.2010.09.003 CrossRefGoogle ScholarPubMed
Stoecklein, K. S., Ortega, M. S., Spate, L. D., Murphy, C. N. and Prather, R. S. (2021). Improved cryopreservation of in vitro produced bovine embryos using FGF2, LIF, and IGF1. PLOS ONE, 16(2), e0243727. doi: 10.1371/journal.pone.0243727 CrossRefGoogle ScholarPubMed
Sun, Y. L., Ping, Z. G., Li, C. J., Sun, Y. F., Yi, K. L., Chen, L., Li, X. Y., Wang, X. L. and Zhou, X. (2011). Comparative proteomic analysis of follicular fluids from normal and cystic follicles in sows. Reproduction in Domestic Animals, 46(5), 889895. doi: 10.1111/j.1439-0531.2011.01760.x CrossRefGoogle ScholarPubMed
Tatemoto, H., Muto, N., Sunagawa, I., Shinjo, A. and Nakada, T. (2004). Protection of porcine oocytes against cell damage caused by oxidative stress during in vitro maturation: Role of superoxide dismutase activity in porcine follicular fluid. Biology of Reproduction, 71(4), 11501157. doi: 10.1095/biolreprod.104.029264 CrossRefGoogle ScholarPubMed
Teplitz, G. M., Lorenzo, M. S., Maruri, A., Cruzans, P. R., Carou, M. C. and Lombardo, D. M. (2020). Coculture of porcine cumulus–oocyte complexes with porcine luteal cells during IVM: Effect on oocyte maturation and embryo development. Reproduction, Fertility, and Development, 32(16), 12501259. doi: 10.1071/RD20117 CrossRefGoogle ScholarPubMed
Tian, H., Qi, Q., Yan, F. and Hou, J. (2021). Enhancing the developmental competence of prepubertal lamb oocytes by supplementing the in vitro maturation medium with sericin and the fibroblast growth factor 2-leukemia inhibitory factor-insulin-like growth factor 1 combination. Theriogenology, 159(1), 1319 CrossRefGoogle ScholarPubMed
Uhm, S. J., Gupta, M. K., Yang, J. H., Chung, H. J., Min, T. S. and Lee, H. T. (2010). Epidermal growth factor can be used in lieu of follicle-stimulating hormone for nuclear maturation of porcine oocytes in vitro . Theriogenology, 73(8), 10241036. doi: 10.1016/j.theriogenology.2009.11.029 CrossRefGoogle ScholarPubMed
Valleh, M. V., Zandi, N. K., Rasmussen, M. A. and Hyttel, P. (2017). Optimal doses of EGF and GDNF act as biological response modifiers to improve porcine oocyte maturation and quality. Zygote, 25(4), 423433. doi: 10.1017/S0967199417000181 CrossRefGoogle ScholarPubMed
Weaver, A. C., Kelly, J. M., Kind, K. L., Gatford, K. L., Kennaway, D. J., Herde, P. J. and van Wettere, W. H. (2013). Oocyte maturation and embryo survival in nulliparous female pigs (gilts) is improved by feeding a lupin-based high-fibre diet. Reproduction, Fertility, and Development, 25(8), 12161223. doi: 10.1071/RD12329 CrossRefGoogle ScholarPubMed
Yoshioka, K., Suzuki, C. and Onishi, A. (2008). Defined system for in vitro production of porcine embryos using a single basic medium. Journal of Reproduction and Development, 54(3), 208213. doi: 10.1262/jrd.20001 CrossRefGoogle ScholarPubMed
Yuan, Y., Spate, L. D., Redel, B. K., Tian, Y., Zhou, J., Prather, R. S. and Roberts, R. M. (2017). Quadrupling efficiency in production of genetically modified pigs through improved oocyte maturation. Proceedings of the National Academy of Sciences of the United States of America, 114(29), E5796E5804. doi: 10.1073/pnas.1703998114 Google ScholarPubMed
Zheng, J., Wen, Y., Song, Y., Wang, K., Chen, D. B. and Magness, R. R. (2008). Activation of multiple signaling pathways is critical for fibroblast growth factor 2- and vascular endothelial growth factor-stimulated ovine fetoplacental endothelial cell proliferation. Biology of Reproduction, 78(1), 143150. doi: 10.1095/biolreprod.107.064477 CrossRefGoogle ScholarPubMed
Zijlstra, C., Kidson, A., Schoevers, E. J., Daemen, A. J., Tharasanit, T., Kuijk, E. W., Hazeleger, W., Ducro-Steverink, D. W., Colenbrander, B. and Roelen, B. A. (2008). Blastocyst morphology, actin cytoskeleton quality and chromosome content are correlated with embryo quality in the pig. Theriogenology, 70(6), 923935. doi: 10.1016/j.theriogenology.2008.05.055 CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Effect of each treatment on the proportion of mature oocytes. Mean of the effects of the studied supplements on nuclear maturation of prepubertal gilt oocytes after 44 h of culture. Error bars show the 95% confidence interval. Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.

Figure 1

Table 1. Distribution of cortical granules (cytoplasmic maturation) in oocytes after 44 h of in vitro maturation

Figure 2

Figure 2. Cortical granule (CG) distribution in oocytes after IVM. GV: Homogenous distribution of CGs throughout the cytoplasm in a germinal vesicle (GV) stage oocyte denoted with the arrow. MI: Peripheral distribution on metaphase I (MI) stage oocyte, CGs begin to locate subjacent to the oolemma. MII: Complete distribution on a mature oocyte (metaphase II, MII), CGs are concentrated at the periphery of the cytoplasm and around the polar body (PB). CGs stained in green (FITC-PNA), DNA stained in blue (Hoechst), ×200 total magnification. Each column displays an individual oocyte; the top row displays a combined image, with the subsequent images showing individual fluorophores (FITC or Hoechst).

Figure 3

Figure 3. Proportion of blastocyst formation after the different treatments on in vitro maturation. Mean of the effects of the studied supplements on blastulation rates after matured prepubertal gilt oocytes were fertilized. Error bars show the 95% confidence interval. Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.

Figure 4

Figure 4. Pig blastocyst morphology. (A–C) Images taken by the use of epifluorescence microscopy at ×200 total magnification. Nuclei in blue (Hoechst staining). (D–F) Phase-contrast optical microscopy images at ×200 total magnification. (A, D) Blastocysts of excellent morphology (grade 1); (B, E) blastocysts of good morphology (grade 2); (C, F) blastocysts of poor morphology (grade 3). Arrows denotate the inner cell mass (ICM) of the blastocysts.

Figure 5

Table 2. Blastocyst quality after the different treatments on in vitro maturation

Figure 6

Figure 5. Average number of cells per blastocyst. Mean of the effects of the different combinations on the number of cells per blastocyst after matured prepubertal gilt oocytes were fertilized. Error bars show the 95% confidence interval (N = 73). Groups did not differ (P > 0.05). FLI, combination of the cytokines FGF2, LIF, and IGF1; sFF, sow follicular fluid.