Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3447 2024-01-16 13:40:46 |
2 format change Meta information modification 3447 2024-01-17 01:59:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mo, L.; Ma, J.; Xiong, Y.; Xiong, X.; Lan, D.; Li, J.; Yin, S. Factors Influencing Yak Oocytes Maturation and Developmental Competence. Encyclopedia. Available online: https://encyclopedia.pub/entry/53902 (accessed on 16 May 2024).
Mo L, Ma J, Xiong Y, Xiong X, Lan D, Li J, et al. Factors Influencing Yak Oocytes Maturation and Developmental Competence. Encyclopedia. Available at: https://encyclopedia.pub/entry/53902. Accessed May 16, 2024.
Mo, Luoyu, Jun Ma, Yan Xiong, Xianrong Xiong, Daoliang Lan, Jian Li, Shi Yin. "Factors Influencing Yak Oocytes Maturation and Developmental Competence" Encyclopedia, https://encyclopedia.pub/entry/53902 (accessed May 16, 2024).
Mo, L., Ma, J., Xiong, Y., Xiong, X., Lan, D., Li, J., & Yin, S. (2024, January 16). Factors Influencing Yak Oocytes Maturation and Developmental Competence. In Encyclopedia. https://encyclopedia.pub/entry/53902
Mo, Luoyu, et al. "Factors Influencing Yak Oocytes Maturation and Developmental Competence." Encyclopedia. Web. 16 January, 2024.
Factors Influencing Yak Oocytes Maturation and Developmental Competence
Edit

The yak (Bos grunniens) is a unique breed living on the Qinghai–Tibet Plateau and its surrounding areas, providing locals with a variety of vital means of living and production. However, the yak has poor sexual maturity and low fertility. High-quality mature oocytes are the basis of animal breeding technology. In vitro culturing of oocytes and embryo engineering technology have been applied to yak breeding. 

yak oocyte in vitro maturation embryo development

1. Introduction

The yak is a unique breed that lives in the Qinghai–Tibet Plateau and offers various necessities for local people’s survival and production. However, the reproductive performance of the yak is low. Yaks are generally calved every other year, with one in two years or two in three years, and the average annual reproductive rate for female yaks is less than 60% [1][2]. The yak has low reproductive performance due to various factors, including seasonal breeding, delayed puberty, and a low frequency of estrus [2][3][4][5].
In order to improve the reproductive performance of yaks, assisted reproductive procedures such as in vitro fertilization (IVF), somatic cell nuclear transfer (SCNT), and embryo transfer (ET) have been widely used in yak breeding [6][7][8][9]. These technologies cannot be implemented without high-quality mature oocytes. The process of oogenesis can be divided into three main stages: The first stage is the proliferation stage. This stage begins with the migration of primordial germ cells to the embryonic reproductive ridge that has not yet begun to differentiate. At this time, the female germ cells are called oogonia, and the oogonia carry out mitosis and proliferate continuously. In the middle and late stages of embryonic development, some oogonia begin to accumulate nutrients, grow, and undergo the first meiosis (MI). These cells are called primary oocytes. Primary oocytes go through the leptotene, zygotene, and pachytene stages, and finally arrest in the diplotene stage. The second stage is the growth stage, in which the volume of oocytes increases significantly, the number and shape of organelles changes greatly, and many nutrients, such as protein, carbohydrates, and lipid droplets are accumulated in the cytoplasm. The third stage is the maturity stage [10]. The oocyte completes its growth and approaches the size of the mature oocyte in volume. At this time, the arrested oocyte has an intact nuclear envelope known as the germinal vesicle (GV). Upon sexual maturity, primary oocytes resume meiosis by overcoming the effect of oocyte maturation inhibitors secreted by granulosa cells under the influence of luteinizing hormone (LH) peaks. After that, a series of changes, such as germinal vesicle break down (GVBD), chromosome aggregation, and uniform distribution of organelles in the cytoplasm occurs, and the primary oocyte subsequently completes the first meiosis, produces a secondary oocyte with half the number of chromosomes, and extrudes a first polar body. Then, the oocyte arrests again in the second meiotic metaphase (MII) and waits for fertilization. Sperm penetration causes the second polar body to extrude and the formation of a diploid fertilized egg, initiating the development of the fertilized egg, cleavage, and blastocyst formation [11][12][13].

2. Endogenous Factor

2.1. Dynamic Transcripts and Protein Changes

The maturation and development of oocytes are complicated processes involving the regulation of various genes and proteins [14][15]. Dynamic changes of transcripts and proteins were observed during the development and maturation of yak oocytes [16][17]. Pei Jie et al. constructed the molecular structure of yak ovarian cortex cells using single-cell RNA sequencing (scRNA-seq). They identified the molecular features and biological functions of different cell populations. Differentially expressed genes (DEG) between oocytes and other types of ovarian cells were mainly enriched in cell cycle transition, DNA repair, and chromosome segregation processes, according to Gene Ontology (GO) enrichment analysis. Gene expression specificity testing indicated that the characterized genes CENPF, TOP2A, MIS18BP1, FST, and INHA were highly expressed in yak oocytes. The FST and TOP2A genes could be considered as the molecular features of yak oocytes within primordial follicles. They also discovered that the yak oocytes regulated the other types of ovarian cells primarily via the interaction between the ligands FAM3, INHA, and JAG1 with their corresponding receptors. However, the endothelial, epithelial, and granulosa cells regulated the oocytes principally via the BMP family [18].

2.2. Epigenetic Regulations

Epigenetic regulation is a kind of regulation of gene expression by changing non-gene sequence, mainly including DNA methylation, histone modification (acetylation, methylation, phosphorylation, etc.), and regulation of non-coding RNA, etc., which is a hot research topic in different fields of biology [19][20].

2.3. G Protein-Coupled Receptor 50 (GPR50)

GPR50 is an orphan G protein-coupled receptor on the X chromosome [21]. Previous research revealed that GPR50 was strongly expressed in yak brain, ovary, and testis tissues, implying that GPR50 might have a function in reproductive development [22]. Yao Ying et al. found that the GPR50 protein was centrally expressed in the membrane during the germinal vesicle (GV) phase of yak oocytes, with the highest GPR50 expression level during the MII phase [22]. Based on these observations, researchers investigated the impacts of GPR50 knockdown and overexpression on yak oocytes. The results indicated that the GPR50 knockdown significantly reduced the oocyte maturation rate and the polarbody excretion rate, while GPR50 overexpression exerted no significant influence on the excretion rate and maturity level of the yak oocytes, suggesting that GPR50 might play a crucial role in yak oocyte maturation in vitro [23].

3. Exogenous Factor

3.1. Growth Factor

Growth factors are a class of peptides that regulate multiple effects, such as cell growth and other cellular functions, by binding to specific, high-affinity cell membrane receptors. They play critical roles in resuming oocyte meiosis, oocyte maturation, and follicular development [24][25]. Moreover, growth factors can significantly improve the development ability of embryos and promote blastocyst formation, which has great biological effects on the development processes and different developmental periods of mammalian embryos [26][27][28].
Several important growth factors, such as epidermal growth factor (EGF), insulin-like growth factor I (IGF-1), fibroblast growth factor 10 (FGF10), and leukemia inhibitory factor (LIF) are essential for oocyte maturation and embryo development. EGF can promote cell proliferation, differentiation, and mammalian oocyte maturation [24][29][30][31]. Ma Li et al. discovered that supplementation with 40 μg/mL EGF could significantly improve oocyte maturation and the development ability of parthenogenetic embryos [32]. Pan Yangyang, et al. found that the medium supplemented with 100 ng/mL EGF could significantly increase the yak COC maturation rate, and cleavage and blastocyst rates after fertilization. This might be caused by the inhibition of EGF on the expression of the pro-apoptosis gene BAX and the promotion of EGF on the expression of anti-apoptosis genes BI-1 [33].
IGF-1 belongs to the insulin-like growth factor family [34], which is involved in mediating cellular proliferation, differentiation, and apoptosis, and plays a critical role in mammals’ growth and development [35]. Pan Yangyang et al. discovered that adding 100 ng/mL IGF-1 to the culture medium significantly increased the yak oocyte maturation rate in vitro and the cleavage and blastocyst rates of chemically activated embryos. Further study proved that this result was due to the up-regulation of IGF-1-induced cold-inducible RNA-binding protein (CIRP) [36].
FGF10 is a paracrine fibroblast growth factor involved in numerous biological processes, including embryonic development, cell growth, morphogenesis, tissue repair, tumor growth, and invasion [37][38][39][40]. It is also involved in follicle development and oocyte maturation in various mammals [41][42]. Pan Yangyang et al. found that adding 5 ng/mL of FGF10 to the culture medium of yak COCs improved the yak oocyte maturation rate and fertilization ability. This positive effect was achieved via the up-regulation of FGF10 on CD9, CD81, DNMT1 and DNMT3B expressions in COCs to optimize sperm–egg interaction and DNA methylation during fertilization [6].
LIF is a potent cytokine in the IL-6 family of cytokines [43]. Zhao Tian et al. indicated that adding LIF during the IVM of yak oocytes improved oocyte quality, maturation competence, blastocyst quality, and oocyte development. The addition of LIF (50 ng/mL) to the maturation medium could increase the maturation rate and significantly lower ROS generation and the apoptosis levels of oocytes by increasing the mRNA transcription levels of anti-apoptotic and antioxidant-related genes BCL2, CAPASE3, SURVIVIN, SOD2 and GPX4 in yak oocytes. Furthermore, blastocysts formed from 50 ng/mL LIF-treated oocytes had higher total cell numbers and lower apoptosis rates than the control group [44].

3.2. Antioxidants

ROS are small molecules produced by biological aerobic metabolism, an include superoxide, peroxide, and oxygen radicals [45]. Reactive oxygen molecules are chemically reactive due to extra-nuclear unpaired electrons. Excessive ROS attack intracellular small molecules, such as lipids, proteins, and nucleic acids, leading to DNA degradation in the nucleus and mitochondria, causing intracellular protein denaturation, inactivating some important enzymes, inducing cellular plasma peroxidation, and ultimately triggering cell apoptosis [46]. High ROS levels accelerated the oocyte senescence, reduced oocyte quality, and caused oocyte apoptosis [47][48].
Vitamin A is an indispensable nutrient that regulates physiological processes such as reproduction, embryonic development, vision, growth, cell differentiation, and proliferation [49]. Vitamin A can be oxidized to retinoic acid (RA) via oxidation reactions, and RA functions as a gene expression regulator [50]. Vitamin A regulates oocyte maturation via typical and atypical signaling pathways [51][52]. According to studies, adding 2 μM Vitamin A to yak oocytes in vitro maturation medium significantly increased the rate of IVM and parthenogenetic activation (PA) embryo cleavage rate. The expressions of STRA8, RARA, and RXRA were highest in the MII stage compared with those in the GV and MI stages under the treatment of 2 μM Vitamin A. Additionally, the mRNA expressions of several genes in the typical signaling pathway, including RXRA, RARA, and STRA8, were significantly higher than those of MEK and MEK1, which were node genes of the atypical signaling pathway. These results suggested that RA was mainly dependent on the typical signaling pathway for the yak oocyte development in vitro [53][54].
Vitamin C (ascorbic acid) is a strong water-soluble antioxidant that can catalyze the reduction of oxidized glutathione to reduced glutathione [55]. Exposure of yak oocytes to 1 nM Aflatoxin B1 (AFB1) induced early oocyte apoptosis and increased intracellular ROS levels. It caused incomplete actin and uneven distribution of mitochondria, resulting in decreased quality of mature yak oocytes. However, adding 50 μg/mL Vitamin C to the culture medium protected yak oocytes from the toxic effects of AFB1 exposure. Specifically, 50 μg/mL Vitamin C reduced intra-oocyte ROS levels, repressed early oocyte apoptosis, improved mitochondrial distribution status, and restored actin distribution [56][57].
The addition of antioxidants to oocytes and embryos during in vitro culture is necessary to maintain normal levels of intracellular ROS. Melatonin is a natural endogenous indole hormone produced by the mammalian pineal gland [58]. Since melatonin is fat- and water-soluble, it can easily transfer hydrogen and electrons across cell membranes, directly scavenging free radicals and reducing cellular ROS levels [46]. Peng Wei et al. investigated the effects of melatonin on the IVM of yak oocytes by adding different concentrations of melatonin to the culture medium of yak COCs. They discovered that adding 10−9 M melatonin could significantly increase the oocyte maturation rate, IVF embryo cleavage and blastocysts rates, and GSH content of oocytes and blastocysts. Reductions of ROS levels, mitochondrial protein extent, DNA damage, and cell apoptosis were observed after melatonin treatment. Additionally, 10−9 M melatonin repaired the spindle mismatch and chromosomal abnormalities caused by oxidative stress. The results suggested that 10−9 M melatonin addition could alleviate oxidative stress during the IVM of oocytes and improve the oocyte maturation rate and the developmental ability of subsequent embryos [59][60].

3.3. Microelement

Zinc (Zn) is an essential trace element in mammals that plays an important role in cell growth, proliferation, division, and immunity [61][62]. Xiong Xianrong et al. discovered that adding 2 mg/L zinc sulfate to the IVM medium of yak oocytes increased glutathione (GSH) content, superoxide dismutase (SOD) activity, and the blastocyst rate, and significantly reduced ROS levels. This effect could be achieved by Zn2+ inducing the up-regulation of Zn transporters 3 (ZnT3), Zrt, and Irt-like protein 14 (ZiP14) expressions in yak oocytes [63]. Hu Jiajia et al. obtained similar results in their study. Moreover, they discovered that adding 2 mg/L zinc sulfate significantly increased the expression levels of Solute-linked carrier (SLC30A) and SLC39A family members, including SCL30A3, SLC30A6, SCL30A9, SLC39A6 and SLC39A14 in yak mature oocytes, facilitating the cleavage of the fertilized ovum and blastocyst formation [64]. Feng Yun et al. revealed that adding 0.8 mg/L zinc sulfate could improve the yak oocyte maturation and the efficiency of in vitro fertilization by increasing the antioxidant enzyme gene (SOD1, CAT, TXN1, and PRD1) expression levels, and also up-regulating the cumulus cell expansion related genes (PTX3 and TSG6) in the oocyte [65].
Calcium (Ca) is an important second messenger in cells, and the changes in Ca2+ concentration are closely related to regulating physiological functions by affecting signal transduction [66]. Chen demonstrated that adding Ca2+ at 0.24 mM in yak oocyte culture medium significantly increased the oocyte maturation rate in vitro. The mechanism was that Ca2+ activated the activity of calmodulin-dependent protein kinaseII (CaMKII), increased GSH content, and decreased the ROS level in the oocyte. Ca2+ up-regulated the expressions of BCL-2, EGF, EGFR, and C-FOS, whereas it down-regulated BAX expression [67].
Selenium (Se) is an essential trace element for reproduction, immunity, antioxidant systems, embryonic growth, and other physiological functions [68][69][70]. Xiong Xianrong et al. observed that 2 μg/mL of sodium selenite significantly increased the glutathione peroxidase (GSH-Px) activity in the oocytes and the blastocyst rate of subsequent embryos by adding sodium selenite to the in vitro culture medium of yak COCs. Those effects were achieved by increasing the selenoprotein synthesis-related gene expression levels, including GPX4, SEPP1, RPL22, and CCND1 in oocytes and cumulus cells [71].

3.4. Small Molecule Compounds

Cyclic adenosine monophosphate (cAMP) is the first discovered second messenger. cAMP can regulate the various target genes’ transcription, primarily via protein kinase A (PKA) and its downstream effectors [72]. cAMP plays a key role in maintaining oocyte meiotic arrest and initiating meiotic resumption in mammalian oocytes [73][74]. Previous studies have demonstrated that maintaining cAMP levels in oocytes before oocyte maturation could temporarily repress spontaneous meiotic resumption, thereby improving oocyte developmental competence and subsequent embryonic development [75][76][77][78]. Xiong Xianrong et al. revealed that a supplement with a cAMP activator, cilostazol, would benefit yak oocytes IVM by increasing cAMP and GSH levels and modulating mRNA expression patterns during pre-IVM. Specifically, adding cilostazol to the in vitro maturation medium and pre-IVM for 2 h or 4 h significantly increased the PKA1 and CY3 mRNA expression levels. It also significantly decreased the PDE3A mRNA expression level in yak COCs and blastocysts [79].
Roscovitine and C-type natriuretic peptide (CNP) are two meiotic arrest factors promoting yak oocyte maturation in vitro. Roscovitine is a member of the 2,6,9-trisubstituted purine family and has a structure similar to ATP. Therefore, roscovitine interacts with amino acids in the ATP-binding pocket of the catalytic domain of some Cyclin-dependent kinases (CDK), preventing ATP from binding to CDK, inhibiting CDK activity, and ultimately blocking the cell cycle [80]. Pretreatment of yak COCs with 12.5 μM roscovitine for 6 h followed by conventional IVM improved the quality of yak mature oocytes. Liu Yu counted the cell expansion index (CEI) of ovarian thalamus granulosa cells after treating pre-IVM yak COCs with different concentrations of roscovitine. They discovered that pretreatment with 12.5 μM roscovitine for 6 h significantly increased the CEI of COCs. This treatment significantly reduced the ROS content in oocytes, promoted the uniform distribution of mitochondria, and enhanced the structure of transzonal projections (TZPs), thereby improving the quality of yak oocytes. Furthermore, this treatment significantly up-regulated the mRNA expressions of antioxidant gene SOD2, anti-apoptotic gene BCL-2, and development-related genes GDF9, EGFR, and ZAR1, and significantly down-regulated the mRNA expression of the pro-apoptotic gene BAX [81].
CNP is a natural determinant of meiotic arrest that can maintain gap junction activity and support the key gene expressions essential for oocyte development [82]. Jing Tian counted the number of yak GV oocytes cultured with different concentrations of CNP and discovered that the quality, maturation rate, and blastocyst rate of yak oocytes could be significantly improved by pre-IVM of oocytes at 100 nM CNP for 6 h and IVM for 28 h. This treatment significantly increased TZP and GSH protein expressions and decreased ROS levels in yak oocytes. These effects might be due to CNP significantly promoting the expressions of CNP receptor gene NPR2, anti-apoptotic gene BCL-2, and growth differentiation factor GDF9 in oocytes and blastocysts. CNP suppressed the EGF and its receptor EGFR gene expressions in oocytes while promoting EGF, EGFR, and DNMT1 expressions in blastocysts and repressing the expression of pro-apoptotic gene BAX in blastocysts [83].

3.5. Hormones

Xiao Xiao et al. demonstrated that supplementing IVM medium with 5 μg/mL FSH and 50 IU/mL LH improved the developmental ability of yak oocytes after in vitro fertilization (IVF) [84]. He Honghong et al. studied the effects of different concentrations of FSH concentrations on yak oocytes and revealed that the oocyte maturation rate was highest in the 5 μg/mL FSH treatment group. Further study indicated that FSH might improve yak oocyte development by increasing EGF and EGFR mRNA expression levels, and it might inhibit oocyte apoptosis by increasing antiapoptotic gene BCL-2 expression while reducing pro-apoptotic gene BAX expression [85].
Estradiol (E2) is the most active and predominant maternal estrogen during pregnancy [86]. A supplement of exogenous E2 or promoting endogenous E2 synthesis and secretion can improve oocyte maturation and increase cumulus cell spread during oocyte maturation in various animals [87][88][89][90][91]. Pan Yangyang et al. discovered that adding 10−4 mM endogenous 17β-estradiol to the IVM medium of COCs could increase the cumulus expansion and subsequent oocyte development. These might result from increasing the expressions of cumulus-expansion-related factors (HAS2, PTGS2, and PTX3) and the oocyte-secreted factors (GDF9, FGF10, and BMP15) [92].
Rfamide-related peptides 3 (RFRP-3), a structural and functional homolog of gonadotropin-inhibiting hormone (GnIH), has been proposed as a new breeding inhibitory neurohormonal peptide that plays a crucial role in the reproductive axis across various species [93][94].
Cytochrome P450arom (CYP19A1) is the key enzyme for gonadal hormone synthesis in most animals [95]. CYP19A1 up-regulated the endogenous E2 level and enhanced the developmental ability of yak oocytes. Specifically, the treatment of CYP19A1 activator AFB1 up-regulated the endogenous E2 level and increased the rates of IVM and blastocysts, while decreasing the E2 level. IVM and blastocyst rates were observed in the CYP19A1 inhibitor BPA treatment group, which implied that CYP19A1 played an essential role in oocyte and embryo development of yaks [96].

3.6. Platelet-Activating Factor (PAF)

PAF is an acetylated glycerol signaling phospholipid important physiological regulator in reproduction [97]. PAF exerts its actions via activating specific PAF receptors (PAF-R) in cells and tissues of the female reproductive tract [98]. Wang Qin explored the role of PAF in the maturation of yak oocytes and early embryonic development by adding different concentrations of PAF to the maturation medium of COCs in vitro. The results showed that 10−7 mol/L PFA significantly increased the maturation, cleavage, and blastocyst rates of yak oocytes by regulating BAX, BCL-2, EGF, EGFR, C-FOS, OCT-4, and NANOG gene expressions [99][100].

4. Environment Factor

4.1. Temperature

Pang Bo et al. investigated the effects of ovarian preservation temperature and the culture methods on the maturation rate of yak oocytes in vitro, and the results showed that preserving the ovaries from 20 °C to 25 °C could improve the oocyte maturation rate in vitro [101]. Ma Li et al. preserved fresh yak ovaries in saline at different temperatures (15–20 °C; 25–30 °C; 35–40 °C). The results indicated that 25–30 °C was the optimal temperature for the ovarian transport of yaks. The maturation rate of oocytes, eight-cell embryo formation, and blastocyst rates of IVM and parthenogenetic activation (PA) embryos were significantly higher than those of other groups within this temperature range [102].

4.2. Oxygen

Oxygen (O2) is vital to maintain and complete oocyte maturation and embryonic development. Changes in oxygen concentration during oocyte maturation in vitro affect nuclear DNA methylation, intracellular reactive oxygen species (ROS) levels, and cellular aging [47][103]. Low oxygen levels are the naturally preferred microenvironment for most processes during early development and mainly drive proliferation [104][105]. Several studies proved that culturing oocytes and embryos under low oxygen conditions improved their developmental capacity [106][107][108].
Li Ruizhe compared the expression differences in the transcriptome of yak oocytes at 5% and 20% oxygen concentrations and revealed that the genes up-regulated in the 5% group were mainly involved in hypoxia response, the cell cycle, chromatin conformation and remodeling, and the cytoskeleton, including the WDR26, MKP2K1, MAPK1, TICRR, WAC, EIF4ENIF1, ODC1, CHAMP1, MKI67, MCM10, SFMBT1, PBRM1, KAT8, IQGAP2, EPS8, and RANBP9 genes. The genes up-regulated in the 20% oxygen concentration group, including ACAT1, ATP5MF, AURKAIP1, COX6A1, NDUFA10, NDUFA11, NDUFS7, LYRM7, UQCR10, EIF1AX, RPL13, RPL13A, RPL34, RPLP2, GSTA2, QARS, NOSTRIN, SH3BGRL3, TIMP1, S100A11, and PTX3, were primarily involved in energy metabolism, protein synthesis, redox homeostasis, and oocyte regulation [109]. Additionally, Li Ruizhe et al. discovered that 5% O2 increased the oocyte maturation rate and GSH content, decreased the oocyte ROS level, and improved the quality of PA and IVF blastocysts. The effects were achieved by decreasing the expressions of the antioxidant genes CAT and GPX1, increasing the expression of the metabolism-related gene LDHA, and embryo development-related genes CDX2 and OCT4 in yak IVF blastocysts [109][110].

References

  1. Zhou, J.; Yue, S.; Du, J.; Xue, B.; Wang, L.; Peng, Q.; Zou, H.; Hu, R.; Jiang, Y.; Wang, Z.; et al. Integration of transcriptomic and metabolomic analysis of the mechanism of dietary N-carbamoylglutamate in promoting follicle development in yaks. Front. Vet. Sci. 2022, 9, 946893.
  2. Zi, X.D. Reproduction in female yaks (Bos grunniens) and opportunities for improvement. Theriogenology 2003, 59, 1303–1312.
  3. Ma, J.; Shah, A.M.; Wang, Z.; Hu, R.; Zou, H.; Wang, X.; Cao, G.; Peng, Q.; Xue, B.; Wang, L.; et al. Comparing the gastrointestinal barrier function between growth-retarded and normal yaks on the Qinghai-Tibetan Plateau. PeerJ 2020, 8, e9851.
  4. Yang, X.; Ma, J.; Mo, L.; Xiong, Y.; Xiong, X.; Lan, D.; Fu, W.; Yin, S. Molecular cloning and characterization of STC1 gene and its functional analyses in yak (Bos grunniens) cumulus granulosa cells. Theriogenology 2023, 208, 185–193.
  5. Yin, S.; Zhou, J.; Yang, L.; Yuan, Y.; Xiong, X.; Lan, D.; Li, J. Identification of microRNA transcriptome throughout the lifespan of yak (Bos grunniens) corpus luteum. Anim. Biotechnol. 2023, 34, 143–155.
  6. Pan, Y.; Wang, M.; Baloch, A.R.; Zhang, Q.; Wang, J.; Ma, R.; Xu, G.; Kashif, J.; Wang, L.; Fan, J.; et al. FGF10 enhances yak oocyte fertilization competence and subsequent blastocyst quality and regulates the levels of CD9, CD81, DNMT1, and DNMT3B. J. Cell. Physiol. 2019, 234, 17677–17689.
  7. Niu, H.R.; Zi, X.D.; Xiao, X.; Xiong, X.R.; Zhong, J.C.; Li, J.; Wang, L.; Wang, Y. Developmental competence of frozen-thawed yak (Bos grunniens) oocytes followed by in vitro maturation and fertilization. Cryobiology 2014, 68, 152–154.
  8. Li, Y.; Dai, Y.; Du, W.; Zhao, C.; Wang, L.; Wang, H.; Liu, Y.; Li, R.; Li, N. In vitro development of yak (Bos grunniens) embryos generated by interspecies nuclear transfer. Anim. Reprod. Sci. 2007, 101, 45–59.
  9. Li, Y.; Li, S.; Dai, Y.; Du, W.; Zhao, C.; Wang, L.; Wang, H.; Li, R.; Liu, Y.; Wan, R.; et al. Nuclear reprogramming in embryos generated by the transfer of yak (Bos grunniens) nuclei into bovine oocytes and comparison with bovine-bovine SCNT and bovine IVF embryos. Theriogenology 2007, 67, 1331–1338.
  10. Das, D.; Arur, S. Regulation of oocyte maturation: Role of conserved ERK signaling. Mol. Reprod. Dev. 2022, 89, 353–374.
  11. Yin, S.; Jiang, X.; Jiang, H.; Gao, Q.; Wang, F.; Fan, S.; Khan, T.; Jabeen, N.; Khan, M.; Ali, A.; et al. Histone acetyltransferase KAT8 is essential for mouse oocyte development by regulating reactive oxygen species levels. Development 2017, 144, 2165–2174.
  12. Sen, A.; Caiazza, F. Oocyte maturation: A story of arrest and release. Front. Biosci. (Schol. Ed.) 2013, 5, 451–477.
  13. Duncan, F.E.; Gerton, J.L. Mammalian oogenesis and female reproductive aging. Aging 2018, 10, 162–163.
  14. Strączyńska, P.; Papis, K.; Morawiec, E.; Czerwiński, M.; Gajewski, Z.; Olejek, A.; Bednarska-Czerwińska, A. Signaling mechanisms and their regulation during in vivo or in vitro maturation of mammalian oocytes. Reprod. Biol. Endocrinol. 2022, 20, 37.
  15. Gegenfurtner, K.; Flenkenthaler, F.; Fröhlich, T.; Wolf, E.; Arnold, G.J. The impact of transcription inhibition during in vitro maturation on the proteome of bovine oocytes. Biol. Reprod. 2020, 103, 1000–1011.
  16. Andreu-Vieyra, C.; Lin, Y.N.; Matzuk, M.M. Mining the oocyte transcriptome. Trends Endocrinol. Metab. 2006, 17, 136–143.
  17. Rodriguez, K.F.; Farin, C.E. Gene transcription and regulation of oocyte maturation. Reprod. Fertil. Dev. 2004, 16, 55–67.
  18. Pei, J.; Xiong, L.; Guo, S.; Wang, X.; Bao, P.; Wu, X.; Yan, P.; Guo, X. A single-cell transcriptomic atlas characterizes cell types and their molecular features in yak ovarian cortex. Faseb J. 2023, 37, e22718.
  19. Feinberg, A.P. The Key Role of Epigenetics in Human Disease Prevention and Mitigation. N. Engl. J. Med. 2018, 378, 1323–1334.
  20. Zhang, Y.; Sun, Z.; Jia, J.; Du, T.; Zhang, N.; Tang, Y.; Fang, Y.; Fang, D. Overview of Histone Modification. Adv. Exp. Med. Biol. 2021, 1283, 1–16.
  21. Drew, J.E.; Barrett, P.; Mercer, J.G.; Moar, K.M.; Canet, E.; Delagrange, P.; Morgan, P.J. Localization of the melatonin-related receptor in the rodent brain and peripheral tissues. J. Neuroendocrinol. 2001, 13, 453–458.
  22. Yao, Y.; Chen, Y.; Xiong, X.R.; Chai, Z.X.; Ji, W.H.; Lan, D.L.; MIPAM Tserang-donko. Study on the expression and subcellular localization of G Protein-coupled Receptor 50 during in vitro maturation process of yak oocytes. China Anim. Husb. Vet. Med. 2021, 48, 3387–3393.
  23. Chen, Y.; Zeng, R.; Kou, J.; Xiong, X.; Yao, Y.; Fu, W.; Yin, S.; Lan, D. GPR50 participates in and promotes yak oocyte maturation: A new potential oocyte regulatory molecule. Theriogenology 2022, 181, 34–41.
  24. Bezerra, F.T.G.; Paulino, L.; Silva, B.R.; Silva, A.W.B.; Souza Batista, A.L.P.; Silva, J.R.V. Effects of epidermal growth factor and progesterone on oocyte meiotic resumption and the expression of maturation-related transcripts during prematuration of oocytes from small and medium-sized bovine antral follicles. Reprod. Fertil. Dev. 2020, 32, 1190–1199.
  25. Muhammad, T.; Li, M.; Wang, J.; Huang, T.; Zhao, S.; Zhao, H.; Liu, H.; Chen, Z.J. Roles of insulin-like growth factor II in regulating female reproductive physiology. Sci. China Life Sci. 2020, 63, 849–865.
  26. Currin, L.; Glanzner, W.G.; Gutierrez, K.; de Macedo, M.P.; Guay, V.; Baldassarre, H.; Bordignon, V. Optimizing swine in vitro embryo production with growth factor and antioxidant supplementation during oocyte maturation. Theriogenology 2022, 194, 133–143.
  27. Richter, K.S. The importance of growth factors for preimplantation embryo development and in-vitro culture. Curr. Opin. Obstet. Gynecol. 2008, 20, 292–304.
  28. Dadi, T.D.; Li, M.W.; Lloyd, K.C. Decreased growth factor expression through RNA interference inhibits development of mouse preimplantation embryos. Comp. Med. 2009, 59, 331–338.
  29. Yousef, N.A.; Hussein, H.A.S.; Badr, M.R.; Ahmed, A.E. Effect of Epidermal Growth Factors (EGF) on the Maturation and Developmental Competence of Buffalos’ Oocytes and Embryo Stages In Vitro. SVU-Int. J. Vet. Sci. 2018, 1, 85–94.
  30. Yu, Y.; Yan, J.; Li, M.; Yan, L.; Zhao, Y.; Lian, Y.; Li, R.; Liu, P.; Qiao, J. Effects of combined epidermal growth factor, brain-derived neurotrophic factor and insulin-like growth factor-1 on human oocyte maturation and early fertilized and cloned embryo development. Hum. Reprod. 2012, 27, 2146–2159.
  31. Warzych, E.; Peippo, J.; Szydlowski, M.; Lechniak, D. Supplements to in vitro maturation media affect the production of bovine blastocysts and their apoptotic index but not the proportions of matured and apoptotic oocytes. Anim. Reprod. Sci. 2007, 97, 334–343.
  32. Ma, L.; Yuan, N. The effect of in vitro culture time and EGF on the yak oocyte maturation and parthenogenetic embryos in vitro development. J. Southwest Minzu Univ. (Nat. Sci. Ed.) 2013, 39, 865–868.
  33. Pan, Y.Y.; Li, Q.; Cui, Y.; Fan, J.F.; Yang, K.; He, J.F.; Yu, S.J. The expression of EGF and EGFR in yak oocyte and its function on development competence of embryo. Sci. Agric. Sin. 2015, 48, 2439–2448.
  34. Stuard, W.L.; Titone, R.; Robertson, D.M. The IGF/Insulin-IGFBP Axis in Corneal Development, Wound Healing, and Disease. Front. Endocrinol. 2020, 11, 24.
  35. Bailes, J.; Soloviev, M. Insulin-Like Growth Factor-1 (IGF-1) and Its Monitoring in Medical Diagnostic and in Sports. Biomolecules 2021, 11, 217.
  36. Pan, Y.; Cui, Y.; He, H.; Baloch, A.R.; Fan, J.; Xu, G.; He, J.; Yang, K.; Li, G.; Yu, S. Developmental competence of mature yak vitrified-warmed oocytes is enhanced by IGF-I via modulation of CIRP during in vitro maturation. Cryobiology 2015, 71, 493–498.
  37. Liu, L.; Shi, Q.; Wang, K.; Qian, Y.; Zhou, L.; Bellusci, S.; Chen, C.; Dong, N. Fibroblast growth factor 10 protects against particulate matter-induced lung injury by inhibiting oxidative stress-mediated pyroptosis via the PI3K/Akt/Nrf2 signaling pathway. Int. Immunopharmacol. 2022, 113, 109398.
  38. Itoh, N. FGF10: A multifunctional mesenchymal-epithelial signaling growth factor in development, health, and disease. Cytokine Growth Factor Rev. 2016, 28, 63–69.
  39. Upadhyay, D.; Panduri, V.; Kamp, D.W. Fibroblast growth factor-10 prevents asbestos-induced alveolar epithelial cell apoptosis by a mitogen-activated protein kinase-dependent mechanism. Am. J. Respir. Cell Mol. Biol. 2005, 32, 232–238.
  40. Jiang, T.; Hu, W.; Zhang, S.; Ren, C.; Lin, S.; Zhou, Z.; Wu, H.; Yin, J.; Tan, L. Fibroblast growth factor 10 attenuates chronic obstructive pulmonary disease by protecting against glycocalyx impairment and endothelial apoptosis. Respir. Res. 2022, 23, 269.
  41. Chaves, R.N.; Lima-Verde, I.B.; Celestino, J.J.; Duarte, A.B.; Alves, A.M.; Matos, M.H.; Campello, C.C.; Name, K.P.; Báo, S.N.; Buratini, J., Jr.; et al. Fibroblast growth factor-10 maintains the survival and promotes the growth of cultured goat preantral follicles. Domest. Anim. Endocrinol. 2010, 39, 249–258.
  42. Du, S.; Liu, X.; Deng, K.; Zhou, W.; Lu, F.; Shi, D. The expression pattern of fibroblast growth factor 10 and its receptors during buffalo follicular development. Int. J. Clin. Exp. Pathol. 2018, 11, 4934–4941.
  43. Rose-John, S. Interleukin-6 Family Cytokines. Cold Spring Harb. Perspect. Biol. 2018, 10, a028415.
  44. Zhao, T.; Pan, Y.; Li, Q.; Ding, T.; Niayale, R.; Zhang, T.; Wang, J.; Wang, Y.; Zhao, L.; Han, X.; et al. Leukemia inhibitory factor enhances the development and subsequent blastocysts quality of yak oocytes in vitro. Front. Vet. Sci. 2022, 9, 997709.
  45. Cao, B.; Qin, J.; Pan, B.; Qazi, I.H.; Ye, J.; Fang, Y.; Zhou, G. Oxidative Stress and Oocyte Cryopreservation: Recent Advances in Mitigation Strategies Involving Antioxidants. Cells 2022, 11, 3573.
  46. Jones, D.P. Redefining oxidative stress. Antioxid. Redox Signal 2006, 8, 1865–1879.
  47. Wang, L.; Tang, J.; Wang, L.; Tan, F.; Song, H.; Zhou, J.; Li, F. Oxidative stress in oocyte aging and female reproduction. J. Cell. Physiol. 2021, 236, 7966–7983.
  48. Prasad, S.; Tiwari, M.; Pandey, A.N.; Shrivastav, T.G.; Chaube, S.K. Impact of stress on oocyte quality and reproductive outcome. J. Biomed. Sci. 2016, 23, 36.
  49. Bar-El Dadon, S.; Reifen, R. Vitamin A and the epigenome. Crit. Rev. Food Sci. Nutr. 2017, 57, 2404–2411.
  50. Ikeda, S.; Kitagawa, M.; Imai, H.; Yamada, M. The roles of vitamin A for cytoplasmic maturation of bovine oocytes. J. Reprod. Dev. 2005, 51, 23–35.
  51. Feng, C.W.; Burnet, G.; Spiller, C.M.; Cheung, F.K.M.; Chawengsaksophak, K.; Koopman, P.; Bowles, J. Identification of regulatory elements required for Stra8 expression in fetal ovarian germ cells of the mouse. Development 2021, 148, dev194977.
  52. Manku, G.; Wang, Y.; Merkbaoui, V.; Boisvert, A.; Ye, X.; Blonder, J.; Culty, M. Role of retinoic acid and platelet-derived growth factor receptor cross talk in the regulation of neonatal gonocyte and embryonal carcinoma cell differentiation. Endocrinology 2015, 156, 346–359.
  53. Ji, W.H.; Wang, Y.L.; He, H.H.; Fu, W.; Lan, D.L. Effects of vitamin A on the maturation and subsequent development of yak oocytes in vitro. China Anim. Husb. Vet. Med. 2022, 49, 4707–4714.
  54. Wang, Y.L. A Preliminary Study on the Effect of Vitamin A on the Maturation of Yak Oocytes In Vitro and Its Mechanism of Actin. Master’s Thesis, Southwest Minzu University, Chengdu, China, 2022.
  55. Knight, J.; Madduma-Liyanage, K.; Mobley, J.A.; Assimos, D.G.; Holmes, R.P. Ascorbic acid intake and oxalate synthesis. Urolithiasis 2016, 44, 289–297.
  56. Li, Q. Application of Ascorbic Aicd in the Regulation of DNA Methylation in Yak Oocytes and IVF Embryos. Ph.D. Thesis, Gansu Agricultural University, Lanzhou, China, 2020.
  57. Li, Q.; Zhao, T.; He, H.; Robert, N.; Ding, T.; Hu, X.; Zhang, T.; Pan, Y.; Cui, Y.; Yu, S. Ascorbic acid protects the toxic effects of aflatoxin B(1) on yak oocyte maturation. Anim. Sci. J. 2022, 93, e13702.
  58. Reiter, R.J.; Tan, D.X.; Fuentes-Broto, L. Melatonin: A multitasking molecule. Prog. Brain Res. 2010, 181, 127–151.
  59. Peng, W.; Huang, R.; Shu, S.; Xv, S.R.; Zhang, J. Research of metatonin inhibits oxidative stress and promotes oocyte maturation of yak in vitro. Chin. Qinghai J. Anim. Vet. Sci. 2021, 51, 1–6+66.
  60. Peng, W. Analysis of Ovarian Differences in Cold and Warm Season and the Effects of Melatonin on Oocyte Maturation and Embryo Development of Yak In Vitro. Ph.D. Thesis, Northwest A&F University, Xianyang, China, 2019.
  61. Benazic, S.; Silconi, Z.B.; Jevtovic, A.; Jurisevic, M.; Milovanovic, J.; Mijajlovic, M.; Nikolic, M.; Kanjevac, T.; Potočňák, I.; Samoľová, E.; et al. The Zn(S-pr-thiosal)(2) complex attenuates murine breast cancer growth by inducing apoptosis and G1/S cell cycle arrest. Future Med. Chem. 2020, 12, 897–914.
  62. Bonaventura, P.; Benedetti, G.; Albarède, F.; Miossec, P. Zinc and its role in immunity and inflammation. Autoimmun. Rev. 2015, 14, 277–285.
  63. Xiong, X.; Lan, D.; Li, J.; Lin, Y.; Zi, X. Effects of Zinc Supplementation During In Vitro Maturation on Meiotic Maturation of Oocytes and Developmental Capacity in Yak. Biol. Trace Elem. Res. 2018, 185, 89–97.
  64. Hu, J.J. Effects of Zinc Supplementation on Meiosis Maturation and Development of Yak Oocytes. Master’s Thesis, Southwest Minzu University, Chengdu, China, 2017.
  65. Feng, Y.; Zhao, X.; Ruan, Z.Y.; Shen, P.L.; Shi, D.S.; Lu, F.H. Effects of Zinc on bovine oocytes in vitro maturation and early embryonic development. Chin. J. Anim. Sci. 2019, 55, 14–19.
  66. Clapham, D.E. Calcium signaling. Cell 2007, 131, 1047–1058.
  67. Chen, Z. Mechanism of Ca2+ Regulating Maturation through CaMKⅡ in Yak. Master’s Thesis, Northwest Minzu University, Lanzhou, China, 2022.
  68. Saito, Y.; Yoshida, Y.; Akazawa, T.; Takahashi, K.; Niki, E. Cell death caused by selenium deficiency and protective effect of antioxidants. J. Biol. Chem. 2003, 278, 39428–39434.
  69. Zeng, H. Selenium as an essential micronutrient: Roles in cell cycle and apoptosis. Molecules 2009, 14, 1263–1278.
  70. Avery, J.C.; Hoffmann, P.R. Selenium, Selenoproteins, and Immunity. Nutrients 2018, 10, 1203.
  71. Xiong, X.; Lan, D.; Li, J.; Lin, Y.; Li, M. Selenium supplementation during in vitro maturation enhances meiosis and developmental capacity of yak oocytes. Anim. Sci. J. 2018, 89, 298–306.
  72. Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y.; Hua, H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp. Hematol. Oncol. 2020, 9, 32.
  73. Pan, B.; Li, J. The art of oocyte meiotic arrest regulation. Reprod. Biol. Endocrinol. 2019, 17, 8.
  74. Appeltant, R.; Somfai, T.; Maes, D.; Van Soom, A.; Kikuchi, K. Porcine oocyte maturation in vitro: Role of cAMP and oocyte-secreted factors—A practical approach. J. Reprod. Dev. 2016, 62, 439–449.
  75. Yang, C.R.; Wei, Y.; Qi, S.T.; Chen, L.; Zhang, Q.H.; Ma, J.Y.; Luo, Y.B.; Wang, Y.P.; Hou, Y.; Schatten, H.; et al. The G protein coupled receptor 3 is involved in cAMP and cGMP signaling and maintenance of meiotic arrest in porcine oocytes. PLoS ONE 2012, 7, e38807.
  76. Vaccari, S.; Horner, K.; Mehlmann, L.M.; Conti, M. Generation of mouse oocytes defective in cAMP synthesis and degradation: Endogenous cyclic AMP is essential for meiotic arrest. Dev. Biol. 2008, 316, 124–134.
  77. Ramos Leal, G.; Santos Monteiro, C.A.; Souza-Fabjan, J.M.G.; de Paula Vasconcelos, C.O.; Garcia Nogueira, L.A.; Reis Ferreira, A.M.; Varella Serapião, R. Role of cAMP modulator supplementations during oocyte in vitro maturation in domestic animals. Anim. Reprod. Sci. 2018, 199, 1–14.
  78. Ozturk, S. Molecular determinants of the meiotic arrests in mammalian oocytes at different stages of maturation. Cell Cycle 2022, 21, 547–571.
  79. Xiong, X.R.; Lan, D.L.; Li, J.; Lin, Y.Q.; Li, M.Y. Supplementation of cilostazol during in vitro maturation enhances the meiosis and developmental competence of yak oocytes by influencing cAMP content and mRNA expression. Anim. Reprod. Sci. 2017, 186, 21–30.
  80. Wesierska-Gadek, J.; Kramer, M.P.; Maurer, M. Resveratrol modulates roscovitine-mediated cell cycle arrest of human MCF-7 breast cancer cells. Food Chem. Toxicol. 2008, 46, 1327–1333.
  81. Liu, Y. The Effects of Roscovitine on Granulosa Cell Culture and Oocyte In Vitro Maturation of the Yak. Master’s Thesis, Southwest Minzu University, Chengdu, China, 2022.
  82. Caixeta, F.M.; Sousa, R.V.; Guimarães, A.L.; Leme, L.O.; Sprícigo, J.F.; Netto, S.B.; Pivato, I.; Dode, M.A. Meiotic arrest as an alternative to increase the production of bovine embryos by somatic cell nuclear transfer. Zygote 2017, 25, 32–40.
  83. Jing, T. The Effects of CNP on Oocyte Maturation and Embryo Development of the Yak. Master’s Thesis, Southwest Minzu University, Chengdu, China, 2021.
  84. Xiao, X.; Zi, X.D.; Niu, H.R.; Xiong, X.R.; Zhong, J.C.; Li, J.; Wang, L.; Wang, Y. Effect of addition of FSH, LH and proteasome inhibitor MG132 to in vitro maturation medium on the developmental competence of yak (Bos grunniens) oocytes. Reprod. Biol. Endocrinol. 2014, 12, 30.
  85. He, H.H.; Pan, Y.Y.; Zhang, H.Z.; Li, Q.; Wang, M.; Cui, Y.; Fan, J.F.; Yu, S.J. The effects of Follicle-stimulating Hormone (FSH) on the expression of EGF and EGFR in yak oocytes and apoptpsis. Acta Vet. Zootech. Sin. 2018, 49, 1899–1907.
  86. Krasnow, J.S.; Hickey, G.J.; Richards, J.S. Regulation of aromatase mRNA and estradiol biosynthesis in rat ovarian granulosa and luteal cells by prolactin. Mol. Endocrinol. 1990, 4, 13–21.
  87. Duan, J.X. The Effect and Regulation of Estrogen to Autophagy in Procine Oocyte. Ph.D. Thesis, Northwest A&F University, Xianyang, China, 2021.
  88. Yang, B.X. Study on the Role of Estradiol in Ovine Cumulus-Oocyte Complexes Cultured In Vitro. Master’s Thesis, Inner Mongolia Agricultural University, Hohhot, China, 2021.
  89. Lv, L.H.; Li, F.Z.; Yue, W.B. Effect of different concentrations of estradiol added to maturation fluid on the maturation of goat oocytes. Heilongjiang Anim. Sci. Vet. Med. 2008, 37–38.
  90. Jin, H.X.; Sun, Y.P.; Xin, Z.M.; Su, Y.C.; Guo, Y.H. Effects of 17 beta-estradiol on maturation and embryo development competence of mouse oocytes in vitro. J. Zhengzhou Univ. (Med. Sci.) 2008, 1137–1139.
  91. Xiao, H.W.; Hua, Z.D.; Zhang, L.P.; Ren, H.Y.; Liu, X.M.; Hua, W.J.; Li, L.; Bi, Y.Z.; Zheng, X.M. Effect of 17-beta-estrodiol (E2) in procine oocytes maturation and fertilized embryos’ development in vitro. Hubei Agric. Sci. 2015, 54, 5660–5661+5665.
  92. Pan, Y.; Wang, M.; Wang, L.; Zhang, Q.; Baloch, A.R.; He, H.; Xu, G.; Soomro, J.; Cui, Y.; Yu, S. Estrogen improves the development of yak (Bos grunniens) oocytes by targeting cumulus expansion and levels of oocyte-secreted factors during in vitro maturation. PLoS ONE 2020, 15, e0239151.
  93. Kriegsfeld, L.J.; Gibson, E.M.; Williams, W.P., 3rd; Zhao, S.; Mason, A.O.; Bentley, G.E.; Tsutsui, K. The roles of RFamide-related peptide-3 in mammalian reproductive function and behaviour. J. Neuroendocrinol. 2010, 22, 692–700.
  94. Mamgain, A.; Sawyer, I.L.; Timajo, D.A.M.; Rizwan, M.Z.; Evans, M.C.; Ancel, C.M.; Inglis, M.A.; Anderson, G.M. RFamide-Related Peptide Neurons Modulate Reproductive Function and Stress Responses. J. Neurosci. 2021, 41, 474–488.
  95. Sun, Y.; Wang, M.; Sun, X.J.; Wang, J.L.; Ma, R.; Yu, S.J.; Pan, Y.Y. Differential expression analysis of CYP19A1 during follicular development and oocyte maturation in yaks. Gansu Anim. Husb. Vet. 2021, 51, 37–42.
  96. Wang, L.B.; Wang, M.; Sun, Y.; Chen, R.; Zhang, T.T.; Huang, Z.H.; Zhang, Q.; Yu, S.J.; Pan, Y.Y. CYP19A1 promotes autophagy and early development ability of yak oocytes by regulating the levels of endogenous estradiol. Acta Vet. Zootech. Sin. 2022, 53, 4283–4295.
  97. Harper, M.J. Platelet-activating factor: A paracrine factor in preimplantation stages of reproduction? Biol. Reprod. 1989, 40, 907–913.
  98. Tiemann, U. The role of platelet-activating factor in the mammalian female reproductive tract. Reprod. Domest. Anim. 2008, 43, 647–655.
  99. Wang, Q. The Effects of PAF on Oocyte Maturation and Embryo Development of the Yak. Master’s Thesis, Southwest Minzu University, Chengdu, China, 2019.
  100. Wang, Q.; Xiong, Y.; Zi, X.D. Effects of PFA supplementation during in vitro maturation medium on yak oocyte development competence and gene expression. Acta Vet. Zootech. Sin. 2020, 51, 514–523.
  101. Pang, B.; Zhao, X.Q.; Guo, Z.L.; Qv, L.; Yu, H.H.; Guo, S.C. Influence of different ovary storage temperature on oocyte in vitro maturation and discovery of yak oocyte maturation cultivation methods. Xinjiang Agric. Sci. 2012, 49, 1158–1164.
  102. Ma, L.; Li, Z.Q.; Xiong, X.R. The effect of yak ovarian transport condition on the oocytes quality and embryos in vitro development. J. Southwest Minzu Univ. (Nat. Sci. Ed.) 2013, 39, 1–4.
  103. Bennemann, J.; Grothmann, H.; Wrenzycki, C. Reduced oxygen concentration during in vitro oocyte maturation alters global DNA methylation in the maternal pronucleus of subsequent zygotes in cattle. Mol. Reprod. Dev. 2018, 85, 849–857.
  104. Fathollahipour, S.; Patil, P.; Leipzig, N. Oxygen Regulation in Development: Lessons from Embryogenesis towards Tissue Engineering. Cells Tissues Organs 2018, 205, 350–371.
  105. Lim, M.; Thompson, J.G.; Dunning, K.R. HYPOXIA AND REPRODUCTIVE HEALTH: Hypoxia and ovarian function: Follicle development, ovulation, oocyte maturation. Reproduction 2021, 161, F33–F40.
  106. Belli, M.; Antonouli, S.; Palmerini, M.G.; Bianchi, S.; Bernardi, S.; Khalili, M.A.; Donfrancesco, O.; Nottola, S.A.; Macchiarelli, G. The effect of low and ultra-low oxygen tensions on mammalian embryo culture and development in experimental and clinical IVF. Syst. Biol. Reprod. Med. 2020, 66, 229–235.
  107. Sánchez-Ajofrín, I.; Iniesta-Cuerda, M.; Sánchez-Calabuig, M.J.; Peris-Frau, P.; Martín-Maestro, A.; Ortiz, J.A.; Del Rocío Fernández-Santos, M.; Garde, J.J.; Gutiérrez-Adán, A.; Soler, A.J. Oxygen tension during in vitro oocyte maturation and fertilization affects embryo quality in sheep and deer. Anim. Reprod. Sci. 2020, 213, 106279.
  108. Hashimoto, S.; Minami, N.; Takakura, R.; Yamada, M.; Imai, H.; Kashima, N. Low oxygen tension during in vitro maturation is beneficial for supporting the subsequent development of bovine cumulus-oocyte complexes. Mol. Reprod. Dev. 2000, 57, 353–360.
  109. Li, R.Z. Effects and Mechanisms of Low Oxygen Concentration on In Vitro Maturation and Developmental Competence of Yak Oocytes. Ph.D. Thesis, Gansu Agricultural University, Lanzhou, China, 2021.
  110. Li, R.; Luo, Y.; Xu, J.; Sun, Y.; Ma, Z.; Chen, S. Effects of oxygen concentrations on developmental competence and transcriptomic profile of yak oocytes. Zygote 2020, 28, 459–469.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 84
Revisions: 2 times (View History)
Update Date: 17 Jan 2024
1000/1000