Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

The oocyte: the key player in the success of assisted reproduction technologies

Trudee Fair https://orcid.org/0000-0002-5079-8589 A * and Pat Lonergan A
+ Author Affiliations
- Author Affiliations

A School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland.

* Correspondence to: trudee.fair@ucd.ie

Reproduction, Fertility and Development 36(2) 133-148 https://doi.org/10.1071/RD23164

© 2024 The Author(s) (or their employer(s)). Published by CSIRO Publishing on behalf of the IETS

Abstract

The ovulation of a mature oocyte at metaphase II of meiosis, with optimal potential to undergo fertilisation by a sperm cell, complete meiosis and sustain the switch to mitotic division, and support early embryo development, involves a protracted and disrupted/delayed series of processes. Many of these are targeted for exploitation in vivo, or recapitulation in vitro, by the livestock industry. Reproductive technologies, including AI, multiple ovulation embryo transfer, ovum pick-up, in vitro embryo production, and oestrus and ovulation synchronisation, offer practitioners and producers the opportunity to produce offspring from genetically valuable dams in much greater numbers than they would normally have in their lifetime, while in vitro oocyte and follicle culture are important platforms for researchers to interrogate the physiological mechanisms driving fertility. The majority of these technologies target the ovarian follicle and the oocyte within; thus, the quality and capability of the recovered oocyte determine the success of the reproductive intervention. Molecular and microscopical technologies have grown exponentially, providing powerful platforms to interrogate the molecular mechanisms which are integral to or affected by ART. The development of the bovine oocyte from its differentiation in the ovary to ovulation is described in the light of its relevance to key aspects of individual interventions, while highlighting the historical timeline.

Keywords: assisted reproduction, cattle, embryo, embryo transfer, fertility, follicle, oocyte, technology.

References

Abdulrahman Alrabiah N, Evans ACO, Fahey AG, Cantwell N, Lonergan P, McCormack J, Browne JA, Fair T (2021) Immunological aspects of ovarian follicle ovulation and corpus luteum formation in cattle. Reproduction 162, 209-225.
| Crossref | Google Scholar | PubMed |

Adamiak SJ, Mackie K, Watt RG, Webb R, Sinclair KD (2005) Impact of nutrition on oocyte quality: cumulative effects of body composition and diet leading to hyperinsulinemia in cattle. Biology of Reproduction 73(5), 918-926.
| Crossref | Google Scholar | PubMed |

Adams GP, Pierson RA (1995) Bovine model for study of ovarian follicular dynamics in humans. Theriogenology 43(1), 113-120.
| Crossref | Google Scholar |

Adams GP, Matteri RL, Kastelic JP, Ko JCH, Ginther OJ (1992) Association between surges of follicle-stimulating hormone and the emergence of follicular waves in heifers. Reproduction 94(1), 177-188.
| Crossref | Google Scholar |

Adams GP, Jaiswal R, Singh J, Malhi P (2008) Progress in understanding ovarian follicular dynamics in cattle. Theriogenology 69(1), 72-80.
| Crossref | Google Scholar | PubMed |

Adhikari D, Zheng W, Shen Y, Gorre N, Hämäläinen T, Cooney AJ, Huhtaniemi I, Lan Z-J, Liu K (2010) Tsc/mTORC1 signaling in oocytes governs the quiescence and activation of primordial follicles. Human Molecular Genetics 19, 397-410.
| Crossref | Google Scholar | PubMed |

Albertini DF, Combelles CM, Benecchi E, Carabatsos MJ (2001) Cellular basis for paracrine regulation of ovarian follicle development. Reproduction 121(5), 647-653.
| Crossref | Google Scholar | PubMed |

Albuz FK, Sasseville M, Lane M, Armstrong DT, Thompson JG, Gilchrist RB (2010) Simulated physiological oocyte maturation (SPOM): a novel in vitro maturation system that substantially improves embryo yield and pregnancy outcomes. Human Reproduction 25(12), 2999-3011.
| Crossref | Google Scholar | PubMed |

Aparicio IM, Garcia-Herreros M, O’Shea LC, Hensey C, Lonergan P, Fair T (2011) Expression, regulation, and function of progesterone receptors in bovine cumulus oocyte complexes during in vitro maturation. Biology of Reproduction 84(5), 910-921.
| Crossref | Google Scholar | PubMed |

Assey RJ, Hyttel P, Greve T, Purwantara B (1994) Oocyte morphology in dominant and subordinate follicles. Molecular Reproduction & Development 37, 335-344.
| Crossref | Google Scholar | PubMed |

Baker TG, Hunter RHF (1978) Oogenesis and follicular growth in the cow: implications for superovulation. In ‘Control of reproduction in the cow’. (Ed. JM Sreenan) pp. 34–49. (CEC Publications: Luxembourg).

Baldassarre H (2021) Laparoscopic ovum pick-up followed by in vitro embryo production and transfer in assisted breeding programs for ruminants. Animals 11(1), 216.
| Crossref | Google Scholar |

Barbosa Latorraca L, Galvão A, Kelsey G, D’augero J, Rabaglino MB, Fair T (2023a) 159 Differential gene expression across bovine oocyte growth phase. Reproduction, Fertility and Development 35(2), 207.
| Crossref | Google Scholar |

Barbosa Latorraca L, Galvão A, Kelsey G, Santos Constancia MdF, Augero JM, Fair T (2023b) 041 Single-cell analysis of DNA methylation in growing bovine oocytes. Animal - Science Proceedings 14(3), 455-456.
| Crossref | Google Scholar |

Barlow DP, Bartolomei MS (2014) Genomic imprinting in mammals. Cold Spring Harbor Perspectives in Biology 6(2), a018382.
| Crossref | Google Scholar | PubMed |

Barnes FL, Sirard M-A (2000) Oocyte maturation. Seminars in Reproductive Medicine 18(2), 123-132.
| Crossref | Google Scholar | PubMed |

Baruselli PS, Batista EOS, Vieira LM, Ferreira RM, Guerreiro BG, Bayeux BM, Sales JNS, Souza AH, Gimenes LU (2016) Factors that interfere with oocyte quality for in vitro production of cattle embryos: effects of different developmental & reproductive stages. Animal Reproduction 13, 264-272.
| Crossref | Google Scholar |

Batista EOS, Guerreiro BM, Freitas BG, Silva JCB, Vieira LM, Ferreira RM, Rezende RG, Basso AC, Lopes RNVR, Rennó FP, Souza AH, Baruselli PS (2016) Plasma anti-Müllerian hormone as a predictive endocrine marker to select Bos taurus (holstein) and Bos indicus (nelore) calves for in vitro embryo production. Domestic Animal Endocrinology 54, 1-9.
| Crossref | Google Scholar | PubMed |

Bebbere D, Albertini DF, Coticchio G, Borini A, Ledda S (2021) The subcortical maternal complex: emerging roles and novel perspectives. Molecular Human Reproduction 27(7), gaab043.
| Crossref | Google Scholar |

Ben-Menahem D (2018) Preparation, characterization and application of long-acting FSH analogs for assisted reproduction. Theriogenology 112, 11-17.
| Crossref | Google Scholar | PubMed |

Bergamo LZ, Bonato DV, Bizarro-Silva C, Bonato FGC, González SM, Rossaneis AC, Verri WA, Jr, Morotti F, Seneda MM (2022a) Culture of preantral ovarian follicles of Bos taurus indicus with alpha-lipoic acid. Zygote 30(2), 206-212.
| Crossref | Google Scholar | PubMed |

Bergamo LZ, Bonato DV, Bizarro-Silva C, Bonato FGC, Sanches TK, Cerezetti MB, Rossaneis AC, Verri WA, Jr, Morotti F, Seneda MM (2022b) Follicular development, morphological integrity, and oxidative stress in bovine preantral follicles cultured in vitro with ascorbic acid. Zygote 30, 391-397.
| Crossref | Google Scholar | PubMed |

Bishop TF, Van Eenennaam AL (2020) Genome editing approaches to augment livestock breeding programs. Journal of Experimental Biology 223(Suppl_1), jeb207159.
| Crossref | Google Scholar |

Bisinotto RS, Lean IJ, Thatcher WW, Santos JEP (2015) Meta-analysis of progesterone supplementation during timed artificial insemination programs in dairy cows. Journal of Dairy Science 98(4), 2472-2487.
| Crossref | Google Scholar | PubMed |

Bizarro-Silva C, Santos MM, Gerez JR, González SM, Lisboa LA, Seneda MM (2018) Influence of follicle-stimulating hormone concentrations on the integrity and development of bovine follicles cultured in vitro. Zygote 26(5), 417-423.
| Crossref | Google Scholar | PubMed |

Blondin P, Bousquet D, Twagiramungu H, Barnes F, Sirard M-A (2002) Manipulation of follicular development to produce developmentally competent bovine oocytes. Biology of Reproduction 66(1), 38-43.
| Crossref | Google Scholar | PubMed |

Bourc’his D, Xu G-L, Lin C-S, Bollman B, Bestor TH (2001) Dnmt3l and the establishment of maternal genomic imprints. Science 294(5551), 2536-2539.
| Crossref | Google Scholar | PubMed |

Britt H, Cushman RA, Dechow CD, Dobson H, Humblot P, Hutjens MF, et al. (2021) Review: Perspective on high-performing dairy cows and herds. Animal 15, 100298.
| Crossref | Google Scholar |

Butler ST, Hutchinson IA, Cromie AR, Shalloo L (2014) Applications and cost benefits of sexed semen in pasture-based dairy production systems. Animal 8(1), 165-172.
| Crossref | Google Scholar |

Byskov AG, Skakkebaek NE, Stafanger G, Peters H (1977) Influence of ovarian surface epithelium and rete ovarii on follicle formation. Journal of Anatomy 123(Pt 1), 77-86.
| Google Scholar | PubMed |

Bó GA, Mapletoft RJ (2014) Historical perspectives and recent research on superovulation in cattle. Theriogenology 81(1), 38-48.
| Crossref | Google Scholar | PubMed |

Bó GA, Rogan DR, Mapletoft RJ (2018) Pursuit of a method for single administration of pFSH for superstimulation in cattle: what we have learned. Theriogenology 112, 26-33.
| Crossref | Google Scholar | PubMed |

Carvalho PD, Santos VG, Giordano JO, Wiltbank MC, Fricke PM (2018) Development of fertility programs to achieve high 21-day pregnancy rates in high-producing dairy cows. Theriogenology 114, 165-172.
| Crossref | Google Scholar | PubMed |

Casida LE, Meyer RK, McShan WH, Wisnicky W (1943) Effects of pituitary gonadotropins on the ovaries and the induction of superfecundity in cattle. American Journal of Veterinary Research 4, 76-94.
| Google Scholar |

Chelenga M, Sakaguchi K, Kawano K, Furukawa E, Yanagawa Y, Katagiri S, Nagano M (2022) Low oxygen environment and astaxanthin supplementation promote the developmental competence of bovine oocytes derived from early antral follicles during 8 days of in vitro growth in a gas-permeable culture device. Theriogenology 177, 116-126.
| Crossref | Google Scholar | PubMed |

Chelenga M, Yanagawa Y, Katagiri S, Nagano M (2023) Pre-maturational culture promotes the developmental competence of bovine oocytes derived from an 8-day in vitro growth culture system. Journal of Reproduction and Development 69, 214-217.
| Crossref | Google Scholar | PubMed |

Choi Y, Ballow DJ, Xin Y, Rajkovic A (2008) Lim homeobox gene, Lhx8, is essential for mouse oocyte differentiation and survival. Biology of Reproduction 79, 442-449.
| Crossref | Google Scholar | PubMed |

Clark ZL, Karl KR, Ruebel ML, Latham KE, Ireland JJ (2022a) Excessive follicle-stimulating hormone during ovarian stimulation of cattle may induce premature luteinization of most ovulatory-size follicles. Biology of Reproduction 106(5), 968-978.
| Crossref | Google Scholar | PubMed |

Clark ZL, Ruebel ML, Schall PZ, Karl KR, Ireland JJ, Latham KE (2022b) Follicular hyperstimulation dysgenesis: new explanation for adverse effects of excessive FSH in ovarian stimulation. Endocrinology 163(9), bqac100.
| Crossref | Google Scholar |

Conti M, Hsieh M, Musa Zamah A, Oh JS (2012) Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Molecular and Cellular Endocrinology 356(1-2), 65-73.
| Crossref | Google Scholar | PubMed |

Currin L, Michalovic L, Bellefleur A-M, Gutierrez K, Glanzner W, Schuermann Y, Bohrer RC, Dicks N, da Rosa PR, De Cesaro MP, Lopez R, Grand F-X, Vigneault C, Blondin P, Gourdon J, Baldassarre H, Bordignon V (2017) The effect of age and length of gonadotropin stimulation on the in vitro embryo development of Holstein calf oocytes. Theriogenology 104, 87-93.
| Crossref | Google Scholar | PubMed |

de Figueiredo JR, de Lima LF, Silva JRV, Santos RR (2018) Control of growth and development of preantral follicle: insights from in vitro culture. Animal Reproduction 15(Suppl 1), 648-659.
| Crossref | Google Scholar | PubMed |

de Figueiredo JR, Cadenas J, de Lima LF, Santos RR (2019) Advances in in vitro folliculogenesis in domestic ruminants. Animal Reproduction 16(1), 52-65.
| Crossref | Google Scholar | PubMed |

de Lima MA, Morotti F, Bayeux BM, de Rezende RG, Botigelli RC, De Bem THC, Fontes PK, Nogueira MFG, Meirelles FV, Baruselli PS, da Silveira JC, Perecin F, Seneda MM (2020) Ovarian follicular dynamics, progesterone concentrations, pregnancy rates and transcriptional patterns in Bos indicus females with a high or low antral follicle count. Scientific Reports 10(1), 19557.
| Crossref | Google Scholar | PubMed |

Demetrio DGB, Benedetti E, Demetrio CGB, Fonseca J, Oliveira M, Magalhaes A, Santos RMd (2020) How can we improve embryo production and pregnancy outcomes of Holstein embryos produced in vitro? (12 years of practical results at a California dairy farm). Animal Reproduction 17(3), e20200053.
| Crossref | Google Scholar | PubMed |

Denicol AC, Lopes G, Jr, Mendonça LG, Rivera FA, Guagnini F, Perez RV, et al. (2012) Low progesterone concentration during the development of the first follicular wave reduces pregnancy per insemination of lactating dairy cow. Journal of Dairy Science 95(4), 1794-1806.
| Crossref | Google Scholar |

Desjardins C, Hafs HD (1969) Maturation of bovine female genitalia from birth through puberty. Journal of Animal Science 28(4), 502-507.
| Crossref | Google Scholar | PubMed |

Diaz FA, Gutierrez-Castillo EJ, Foster BA, Hardin PT, Bondioli KR, Jiang Z (2021) Evaluation of seasonal heat stress on transcriptomic profiles and global DNA methylation of bovine oocytes. Frontiers in Genetics 12, 699920.
| Crossref | Google Scholar | PubMed |

Dieci C, Lodde V, Labreque R, Dufort I, Tessaro I, Sirard M-A, Luciano AM (2016) Differences in cumulus cell gene expression indicate the benefit of a pre-maturation step to improve in-vitro bovine embryo production. Molecular Human Reproduction 22(12), 882-897.
| Crossref | Google Scholar | PubMed |

Dieleman SJ, Kruip TA, Fontijne P, de Jong WH, van der Weyden GC (1983) Changes in oestradiol, progesterone and testosterone concentrations in follicular fluid and in the micromorphology of preovulatory bovine follicles relative to the peak of luteinizing hormone. Journal of Endocrinology 97(1), 31-42.
| Crossref | Google Scholar |

Downs SM, Coleman DL, Ward-Bailey PF, Eppig JJ (1985) Hypoxanthine is the principal inhibitor of murine oocyte maturation in a low molecular weight fraction of porcine follicular fluid. Proceedings of the National Academy of Sciences of the United States of America 82(2), 454-458.
| Crossref | Google Scholar |

Driancourt MA (1991) Follicular dynamics in sheep and cattle. Theriogenology 35(1), 55-79.
| Crossref | Google Scholar |

Driancourt MA, Fry RC (1988) Differentiation of ovulatory follicles in sheep. Journal of Animal Science 66(suppl_2), 9-20.
| Google Scholar |

Dubuc K, Marchais M, Gilbert I, Bastien A, Nenonene KE, Khandjian EW, Viger RS, Delbes G, Robert C (2023) Epitranscriptome marks detection and localization of RNA modifying proteins in mammalian ovarian follicles. Journal of Ovarian Research 16(1), 90.
| Crossref | Google Scholar | PubMed |

Eppig JJ (1979) FSH stimulates hyaluronic acid synthesis by oocyte–cumulus cell complexes from mouse preovulatory follicles. Nature 281(5731), 483-484.
| Crossref | Google Scholar | PubMed |

Eppig JJ, O’Brien MJ (1996) Development in vitro of mouse oocytes from primordial follicles. Biology of Reproduction 54(1), 197-207.
| Crossref | Google Scholar | PubMed |

Eppig JJ, Schroeder AC (1989) Capacity of mouse oocytes from preantral follicles to undergo embryogenesis and development to live young after growth, maturation, and fertilization in vitro. Biology of Reproduction 41(2), 268-276.
| Crossref | Google Scholar | PubMed |

Erickson BH (1966a) Development and senescence of the postnatal bovine ovary. Journal of Animal Science 25, 800-805.
| Crossref | Google Scholar | PubMed |

Erickson BH (1966b) Development and radio-response of the prenatal bovine ovary. Reproduction 11(1), 97-105.
| Crossref | Google Scholar |

Fair T (2010) Mammalian oocyte development: checkpoints for competence. Reproduction, Fertility and Development 22(1), 13-20.
| Crossref | Google Scholar | PubMed |

Fair T, Hyttel P, Greve T (1995) Bovine oocyte diameter in relation to maturational competence and transcriptional activity. Molecular Reproduction & Development 42(4), 437-442.
| Crossref | Google Scholar | PubMed |

Fair T, Hyttel P, Greve T, Boland M (1996) Nucleus structure and transcriptional activity in relation to oocyte diameter in cattle. Molecular Reproduction and Development 43(4), 503-512.
| Crossref | Google Scholar |

Fair T, Hulshof SCJ, Hyttel P, Greve T, Boland M (1997a) Oocyte ultrastructure in bovine primordial to early tertiary follicles. Anatomy and Embryology 195(4), 327-336.
| Crossref | Google Scholar | PubMed |

Fair T, Hulshof SCJ, Hyttel P, Greve T, Boland M (1997b) Nucleus ultrastructure and transcriptional activity of bovine oocytes in preantral and early antral follicles. Molecular Reproduction & Development 46(2), 208-215.
| Crossref | Google Scholar | PubMed |

Fair T, Hyttel P, Motlik J, Boland M, Lonergan P (2002) Maintenance of meiotic arrest in bovine oocytes in vitro using butyrolactone I: effects on oocyte ultrastructure and nucleolus function. Molecular Reproduction & Development 62, 375-386.
| Crossref | Google Scholar | PubMed |

Ferreira EM, Vireque AA, Adona PR, Meirelles FV, Ferriani RA, Navarro PAAS (2009) Cytoplasmic maturation of bovine oocytes: structural and biochemical modifications and acquisition of developmental competence. Theriogenology 71(5), 836-848.
| Crossref | Google Scholar | PubMed |

Ferreira RM, Ayres H, Chiaratti MR, Ferraz ML, Araújo AB, Rodrigues CA, Watanabe YF, Vireque AA, Joaquim DC, Smith LC, Meirelles FV, Baruselli PS (2011) The low fertility of repeat-breeder cows during summer heat stress is related to a low oocyte competence to develop into blastocysts. Journal of Dairy Science 94(5), 2383-2392.
| Crossref | Google Scholar | PubMed |

Ferreira RM, Chiaratti MR, Macabelli CH, Rodrigues CA, Ferraz ML, Watanabe YF, Smith LC, Meirelles FV, Baruselli PS (2016) The infertility of repeat-breeder cows during summer is associated with decreased mitochondrial DNA and increased expression of mitochondrial and apoptotic genes in oocytes. Biology of Reproduction 94, 66.
| Crossref | Google Scholar | PubMed |

Fortune JE, Kito S, Wandji SA, Sršeň V (1998) Activation of bovine and baboon primordial follicles in vitro. Theriogenology 49(2), 441-449.
| Crossref | Google Scholar |

Fortune JE, Rivera GM, Evans ACO, Turzillo AM (2001) Differentiation of dominant versus subordinate follicles in cattle. Biology of Reproduction 65, 648-654.
| Crossref | Google Scholar | PubMed |

Fouladi-Nashta AA, Gutierrez CG, Gong JG, Garnsworthy PC, Webb R (2007) Impact of dietary fatty acids on oocyte quality and development in lactating dairy cows. Biology of Reproduction 77(1), 9-17.
| Crossref | Google Scholar | PubMed |

Führer F, Mayr B, Schellander K, Kalat M, Schleger W (1989) Maturation competence and chromatin behaviour in growing and fully grown cattle oocytes. Journal of Veterinary Medicine Series A 36(1-10), 285-291.
| Crossref | Google Scholar |

Gahurova L, Tomizawa S-I, Smallwood SA, Stewart-Morgan KR, Saadeh H, Kim J, Andrews SR, Chen T, Kelsey G (2017) Transcription and chromatin determinants of de novo DNA methylation timing in oocytes. Epigenetics & Chromatin 10, 25.
| Crossref | Google Scholar | PubMed |

Gallardo TD, John GB, Shirley L, Contreras CM, Akbay EA, Haynie JM, Ward SE, Shidler MJ, Castrillon DH (2007) Genomewide discovery and classification of candidate ovarian fertility genes in the mouse. Genetics 177(1), 179-194.
| Crossref | Google Scholar | PubMed |

Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D, Chen ACH, Eckersley-Maslin MA, Ahmad S, Lee YL, Kobayashi T, Ryan D, Zhong J, Zhu J, Wu J, Lan G, Petkov S, Yang J, Antunes L, Campos LS, Fu B, Wang S, Yong Y, Wang X, Xue S-G, Ge L, Liu Z, Huang Y, Nie T, Li P, Wu D, Pei D, Zhang Y, Lu L, Yang F, Kimber SJ, Reik W, Zou X, Shang Z, Lai L, Surani A, Tam PPL, Ahmed A, Yeung WSB, Teichmann SA, Niemann H, Liu P (2019) Establishment of porcine and human expanded potential stem cells. Nature Cell Biology 21(6), 687-699.
| Crossref | Google Scholar | PubMed |

Garcia Barros R, Lodde V, Franciosi F, Luciano AM (2023) A refined culture system of oocytes from early antral follicles promotes oocyte maturation and embryo development in cattle. Reproduction 165(2), 221-233.
| Crossref | Google Scholar | PubMed |

Gomes RG, Lisboa LA, Silva CB, Max MC, Marino PC, Oliveira RL, González SM, Barreiros TRR, Marinho LSR, Seneda MM (2015) Improvement of development of equine preantral follicles after 6 days of in vitro culture with ascorbic acid supplementation. Theriogenology 84(5), 750-755.
| Crossref | Google Scholar | PubMed |

Goszczynski DE, Cheng H, Demyda-Peyrás S, Medrano JF, Wu J, Ross PJ (2019) In vitro breeding: application of embryonic stem cells to animal production. Biology of Reproduction 100(4), 885-895.
| Crossref | Google Scholar | PubMed |

Goszczynski DE, Navarro M, Mutto AA, Ross PJ (2023) Review: Embryonic stem cells as tools for in vitro gamete production in livestock. Animal 17(Suppl 1), 100828.
| Crossref | Google Scholar |

Gougeon A (1996) Regulation of ovarian follicular development in primates: facts and hypotheses. Endocrine Reviews 17(2), 121-155.
| Crossref | Google Scholar | PubMed |

Guerreiro BM, Batista EOS, Vieira LM, Sá Filho MF, Rodrigues CA, Castro Netto A, Silveira CRA, Bayeux BM, Dias EAR, Monteiro FM, Accorsi M, Lopes RNVR, Baruselli PS (2014) Plasma anti-mullerian hormone: an endocrine marker for in vitro embryo production from Bos taurus and Bos indicus donors. Domestic Animal Endocrinology 49, 96-104.
| Crossref | Google Scholar | PubMed |

Guo H, Zhu P, Yan L, Li R, Hu B, Lian Y, Yan J, Ren X, Lin S, Li J, Jin X, Shi X, Liu P, Wang X, Wang W, Wei Y, Li X, Guo F, Wu X, Fan X, Yong J, Wen L, Xie SX, Tang F, Qiao J (2014) The DNA methylation landscape of human early embryos. Nature 511(7511), 606-610.
| Crossref | Google Scholar | PubMed |

Guo F, Yan L, Guo H, Li L, Hu B, Zhao Y, Yong J, Hu Y, Wang X, Wei Y, Wang W, Li R, Yan J, Zhi X, Zhang Y, Jin H, Zhang W, Hou Y, Zhu P, Li J, Zhang L, Liu S, Ren Y, Zhu X, Wen L, Gao YQ, Tang F, Qiao J (2015) The transcriptome and DNA methylome landscapes of human primordial germ cells. Cell 161(6), 1437-1452.
| Crossref | Google Scholar | PubMed |

Gura MA, Relovská S, Abt KM, Seymour KA, Wu T, Kaya H, Turner JMA, Fazzio TG, Freiman RN (2022) TAF4b transcription networks regulating early oocyte differentiation. Development 149(3), dev200074.
| Crossref | Google Scholar |

Gutierrez CG, Ralph JH, Telfer EE, Wilmut I, Webb R (2000) Growth and antrum formation of bovine preantral follicles in long-term culture in vitro. Biology of Reproduction 62(5), 1322-1328.
| Crossref | Google Scholar | PubMed |

Hamazaki N, Kyogoku H, Araki H, Miura F, Horikawa C, Hamada N, Shimamoto S, Hikabe O, Nakashima K, Kitajima TS, Ito T, Leitch HG, Hayashi K (2021) Reconstitution of the oocyte transcriptional network with transcription factors. Nature 589(7841), 264-269.
| Crossref | Google Scholar | PubMed |

Hanna CW, Kelsey G (2021) Features and mechanisms of canonical and noncanonical genomic imprinting. Genes & Development 35(11-12), 821-834.
| Crossref | Google Scholar | PubMed |

Harasimov K, Uraji J, Mönnich EU, Holubcová Z, Elder K, Blayney M, Schuh M (2023) Actin-driven chromosome clustering facilitates fast and complete chromosome capture in mammalian oocytes. Nature Cell Biology 25(3), 439-452.
| Crossref | Google Scholar | PubMed |

Hayashi K, Saitou M (2013) Stepwise differentiation from naïve state pluripotent stem cells to functional primordial germ cells through an epiblast-like state. Methods in Molecular Biology 1074, 175-183.
| Crossref | Google Scholar | PubMed |

Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou M (2011) Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146(4), 519-532.
| Crossref | Google Scholar | PubMed |

Hayashi K, Ogushi S, Kurimoto K, Shimamoto S, Ohta H, Saitou M (2012) Offspring from oocytes derived from in vitro primordial germ cell–like cells in mice. Science 338(6109), 971-975.
| Crossref | Google Scholar | PubMed |

Henricks DM, Dickey JF, Niswender GD (1970) Serum luteinizing hormone and plasma progesterone levels during the estrous cycle and early pregnancy in cows. Biology of Reproduction 2(3), 346-351.
| Crossref | Google Scholar | PubMed |

Hikabe O, Hamazaki N, Nagamatsu G, Obata Y, Hirao Y, Hamada N, Shimamoto S, Imamura T, Nakashima K, Saitou M, Hayashi K (2016) Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature 539(7628), 299-303.
| Crossref | Google Scholar | PubMed |

Hirao Y (2012) Isolation of ovarian components essential for growth and development of mammalian oocytes in vitro. Journal of Reproduction and Development 58(2), 167-174.
| Crossref | Google Scholar | PubMed |

Hiura H, Obata Y, Komiyama J, Shirai M, Kono T (2006) Oocyte growth-dependent progression of maternal imprinting in mice. Genes to Cells 11(4), 353-361.
| Crossref | Google Scholar |

Holubcová Z, Blayney M, Elder K, Schuh M (2015) Error-prone chromosome-mediated spindle assembly favors chromosome segregation defects in human oocytes. Science 348(6239), 1143-1147.
| Crossref | Google Scholar | PubMed |

Hulshof SCJ, Figueiredo JR, Beckers JF, Bevers MM, van den Hurk R (1994) Isolation and characterization of preantral follicles from foetal bovine ovaries. Veterinary Quarterly 16(2), 78-80.
| Crossref | Google Scholar | PubMed |

Hwang YS, Suzuki S, Seita Y, Ito J, Sakata Y, Aso H, Sato K, Hermann BP, Sasaki K (2020) Reconstitution of prospermatogonial specification in vitro from human induced pluripotent stem cells. Nature Communications 11, 5656.
| Google Scholar |

Hyttel P, Greve T, Callesen H (1989) Ultrastructural aspects of oocyte maturation and fertilization in cattle. Journal of Reproduction and Fertility. Supplement 38, 35-47.
| Google Scholar | PubMed |

Hyttel P, Fair T, Callesen H, Greve T (1997) Oocyte growth, capacitation and final maturation in cattle. Theriogenology 47(1), 23-32.
| Crossref | Google Scholar |

Innocenti F, Fiorentino G, Cimadomo D, Soscia D, Garagna S, Rienzi L, Ubaldi FM, Zuccotti M, on behalf of SIERR (2022) Maternal effect factors that contribute to oocytes developmental competence: an update. Journal of Assisted Reproduction and Genetics 39(4), 861-871.
| Crossref | Google Scholar | PubMed |

Ireland JJ, Roche JF (1982) Development of antral follicles in cattle after prostaglandin-induced luteolysis: changes in serum hormones, steroids in follicular fluid, and gonadotropin receptors. Endocrinology 111(6), 2077-2086.
| Crossref | Google Scholar | PubMed |

Ireland JJ, Roche JF (1983a) Development of nonovulatory antral follicles in heifers: changes in steroids in follicular fluid and receptors for gonadotr opins. Endocrinology 112(1), 150-156.
| Crossref | Google Scholar | PubMed |

Ireland JJ, Roche JF (1983b) Growth and differentiation of large antral follicles after spontaneous luteolysis in heifers: changes in concentration of hormones in follicular fluid and specific binding of gonadotropins to follicles. Journal of Animal Science 57(1), 157-167.
| Crossref | Google Scholar | PubMed |

Ireland JJ, Mihm M, Austin E, Diskin MG, Roche JF (2000) Historical perspective of turnover of dominant follicles during the bovine estrous cycle: key concepts, studies, advancements, and terms. Journal of Dairy Science 83(7), 1648-1658.
| Crossref | Google Scholar | PubMed |

John GB, Gallardo TD, Shirley LJ, Castrillon DH (2008) Foxo3 is a PI3K-dependent molecular switch controlling the initiation of oocyte growth. Developmental Biology 321(1), 197-204.
| Crossref | Google Scholar | PubMed |

Jorssen EPA, Langbeen A, Fransen E, Martinez EL, Leroy JLMR, Bols PEJ (2014) Monitoring preantral follicle survival and growth in bovine ovarian biopsies by repeated use of neutral red and cultured in vitro under low and high oxygen tension. Theriogenology 82, 387-395.
| Crossref | Google Scholar | PubMed |

Joshi S, Davies H, Sims LP, Levy SE, Dean J (2007) Ovarian gene expression in the absence of FIGLA, an oocyte-specific transcription factor. BMC Developmental Biology 7, 67.
| Crossref | Google Scholar | PubMed |

Kaneda M, Hirasawa R, Chiba H, Okano M, Li E, Sasaki H (2010) Genetic evidence for Dnmt3a-dependent imprinting during oocyte growth obtained by conditional knockout with Zp3-cre and complete exclusion of Dnmt3b by chimera formation. Genes to Cells 15, 169-179.
| Crossref | Google Scholar | PubMed |

Kato Y, Kaneda M, Hata K, Kumaki K, Hisano M, Kohara Y, Okano M, Li E, Nozaki M, Sasaki H (2007) Role of the Dnmt3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse. Human Molecular Genetics 16(19), 2272-2280.
| Crossref | Google Scholar | PubMed |

Keeble S, Firman RC, Sarver BAJ, Clark NL, Simmons LW, Dean MD (2021) Evolutionary, proteomic, and experimental investigations suggest the extracellular matrix of cumulus cells mediates fertilization outcomes. Biology of Reproduction 105(4), 1043-1055.
| Crossref | Google Scholar | PubMed |

Kim N-H, Cho SK, Choi SH, Kim EY, Park SP, Lim JH (2000) The distribution and requirements of microtubules and microfilaments in bovine oocytes during in vitro maturation. Zygote 8(1), 25-32.
| Crossref | Google Scholar | PubMed |

Kobayashi K, Takagi Y, Satoh T, Hoshi H, Oikawa T (1992) Development of early bovine embryos to the blastocyst stage in serum-free conditioned medium from bovine granulosa cells. In Vitro Cellular & Developmental Biology-Animal 28, 255-259.
| Crossref | Google Scholar |

Kobayashi T, Zhang H, Tang WWC, Irie N, Withey S, Klisch D, Sybirna A, Dietmann S, Contreras DA, Webb R, Allegrucci C, Alberio R, Surani MA (2017) Principles of early human development and germ cell program from conserved model systems. Nature 546(7658), 416-420.
| Crossref | Google Scholar | PubMed |

Kobayashi T, Castillo-Venzor A, Penfold CA, Morgan M, Mizuno N, Tang WWC, Osada Y, Hirao M, Yoshida F, Sato H, Nakauchi H, Hirabayashi M, Surani MA (2021) Tracing the emergence of primordial germ cells from bilaminar disc rabbit embryos and pluripotent stem cells. Cell Reports 37(2), 109812.
| Crossref | Google Scholar | PubMed |

Labrecque R, Lodde V, Dieci C, Tessaro I, Luciano AM, Sirard MA (2015) Chromatin remodelling and histone mRNA accumulation in bovine germinal vesicle oocytes. Molecular Reproduction and Development 82(6), 450-462.
| Crossref | Google Scholar | PubMed |

Landry DA, Bellefleur A-M, Labrecque R, Grand F-X, Vigneault C, Blondin P, Sirard M-A (2016) Effect of cow age on the in vitro developmental competence of oocytes obtained after FSH stimulation and coasting treatments. Theriogenology 86(5), 1240-1246.
| Crossref | Google Scholar | PubMed |

Li L, Baibakov B, Dean J (2008) A subcortical maternal complex essential for preimplantation mouse embryogenesis. Developmental Cell 15(3), 416-425.
| Crossref | Google Scholar | PubMed |

Li HJ, Sutton-McDowall ML, Wang X, Sugimura S, Thompson JG, Gilchrist RB (2016) Extending prematuration with cAMP modulators enhances the cumulus contribution to oocyte antioxidant defence and oocyte quality via gap junctions. Human Reproduction 31(4), 810-821.
| Crossref | Google Scholar | PubMed |

Liu L, Rajareddy S, Reddy P, Jagarlamudi K, Du C, Shen Y, Guo Y, Boman K, Lundin E, Ottander U, Selstam G, Liu K (2007) Phosphorylation and inactivation of glycogen synthase kinase-3 by soluble kit ligand in mouse oocytes during early follicular development. Journal of Molecular Endocrinology 38(1), 137-146.
| Crossref | Google Scholar |

Lodde V, Modina SC, Franciosi F, Zuccari E, Tessaro I, Luciano AM (2009) Localization of DNA methyltransferase-1 during oocyte differentiation, in vitro maturation and early embryonic development in cow. European Journal of Histochemistry 53(4), e24.
| Crossref | Google Scholar |

Lodde V, Franciosi F, Tessaro I, Modina SC, Luciano AM (2013) Role of gap junction-mediated communications in regulating large-scale chromatin configuration remodeling and embryonic developmental competence acquisition in fully grown bovine oocyte. Journal of Assisted Reproduction and Genetics 30, 1219-1226.
| Crossref | Google Scholar | PubMed |

Lonergan P, Fair T (2016) Maturation of oocytes in vitro. Annual Review of Animal Biosciences 4, 255-268.
| Crossref | Google Scholar | PubMed |

Lonergan P, Faerge I, Hyttel PM, Boland M, Fair T (2003) Ultrastructural modifications in bovine oocytes maintained in meiotic arrest in vitro using roscovitine or butyrolactone. Molecular Reproduction and Development 64, 369-378.
| Crossref | Google Scholar | PubMed |

Luciano AM, Sirard M-A (2018) Successful in vitro maturation of oocytes: a matter of follicular differentiation. Biology of Reproduction 98(2), 162-169.
| Crossref | Google Scholar | PubMed |

Luciano AM, Lodde V, Franciosi F, Ceciliani F, Peluso JJ (2010) Progesterone receptor membrane component 1 expression and putative function in bovine oocyte maturation, fertilization, and early embryonic development. Reproduction 140(5), 663-672.
| Crossref | Google Scholar | PubMed |

Luciano AM, Franciosi F, Modina SC, Lodde V (2011) Gap junction-mediated communications regulate chromatin remodeling during bovine oocyte growth and differentiation through cAMP-dependent mechanism(s). Biology of Reproduction 85(6), 1252-1259.
| Crossref | Google Scholar | PubMed |

Luciano AM, Barros RG, Soares ACS, Buratini J, Lodde V, Franciosi F (2021) Recreating the follicular environment: a customized approach for in vitro culture of bovine oocytes based on the origin and differentiation state. Methods in Molecular Biology 2273, 1-15.
| Crossref | Google Scholar | PubMed |

Lucifero D, Mann MRW, Bartolomei MS, Trasler JM (2004) Gene-specific timing and epigenetic memory in oocyte imprinting. Human Molecular Genetics 13(8), 839-849.
| Crossref | Google Scholar | PubMed |

Lucy MC (2011) Growth hormone regulation of follicular growth. Reproduction, Fertility and Development 24(1), 19-28.
| Crossref | Google Scholar | PubMed |

Łuksza M, Queguigner I, Verlhac M-H, Brunet S (2013) Rebuilding MTOCs upon centriole loss during mouse oogenesis. Developmental Biology 382(1), 48-56.
| Crossref | Google Scholar | PubMed |

Luong XG, Daldello EM, Rajkovic G, Yang C-R, Conti M (2020) Genome-wide analysis reveals a switch in the translational program upon oocyte meiotic resumption. Nucleic Acids Research 48(6), 3257-3276.
| Crossref | Google Scholar | PubMed |

Lussier JG, Matton P, Dufour JJ (1987) Growth rates of follicles in the ovary of the cow. Reproduction 81(2), 301-307.
| Crossref | Google Scholar |

Macaulay AD, Gilbert I, Scantland S, Fournier E, Ashkar F, Bastien A, Saadi HAS, Gagné D, Sirard M-A, Khandjian ÉW, Richard FJ, Hyttel P, Robert C (2016) Cumulus cell transcripts transit to the bovine oocyte in preparation for maturation. Biology of Reproduction 94(1), 16.
| Crossref | Google Scholar | PubMed |

Maehama T, Dixon JE (1998) The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. Journal of Biological Chemistry 273(22), 13375-13378.
| Crossref | Google Scholar | PubMed |

Maidarti M, Anderson RA, Telfer EE (2020) Crosstalk between PTEN/PI3K/Akt signalling and DNA damage in the oocyte: implications for primordial follicle activation, oocyte quality and ageing. Cells 9(1), 200.
| Crossref | Google Scholar | PubMed |

Marei WF, Ghafari F, Fouladi-Nashta AA (2012) Role of hyaluronic acid in maturation and further early embryo development of bovine oocytes. Theriogenology 78(3), 670-677.
| Crossref | Google Scholar | PubMed |

Marion GB, Gier HT (1971) Ovarian and uterine embryogenesis and morphology of the non-pregnant female mammal. Journal of Animal Science 32(suppl_1), 24-47.
| Google Scholar | PubMed |

Mayes MA, Sirard MA (2002) Effect of type 3 and type 4 phosphodiesterase inhibitors on the maintenance of bovine oocytes in meiotic arrest. Biology of Reproduction 66(1), 180-184.
| Crossref | Google Scholar |

McLaughlin M, Telfer EE (2010) Oocyte development in bovine primordial follicles is promoted by activin and FSH within a two-step serum-free culture system. Reproduction 139(6), 971-978.
| Crossref | Google Scholar | PubMed |

Medvedev S, Pan H, Schultz RM (2011) Absence of MSY2 in mouse oocytes perturbs oocyte growth and maturation, RNA stability, and the transcriptome. Biology of Reproduction 85(3), 575-583.
| Crossref | Google Scholar | PubMed |

Mihm M, Good TEM, Ireland JLH, Ireland JJ, Knight PG, Roche JF (1997) Decline in serum follicle-stimulating hormone concentrations alters key intrafollicular growth factors involved in selection of the dominant follicle in heifers. Biology of Reproduction 57(6), 1328-1337.
| Crossref | Google Scholar | PubMed |

Mihm M, Curran N, Hyttel P, Knight PG, Boland MP, Roche JF (1999) Effect of dominant follicle persistence on follicular fluid oestradiol and inhibin and on oocyte maturation in heifers. Journal of Reproduction and Fertility 116(2), 293-304.
| Crossref | Google Scholar | PubMed |

Mishra AK, Kumar A, Yadav S, Anand M, Yadav B, Nigam R, Garg SK, Swain DK (2019) Functional insights into voltage gated proton channel (Hv1) in bull spermatozoa. Theriogenology 136, 118-130.
| Crossref | Google Scholar | PubMed |

Nascimento DR, Barbalho EC, Gondim Barrozo L, de Assis EIT, Costa FC, Silva JRV (2023) The mechanisms that control the preantral to early antral follicle transition and the strategies to have efficient culture systems to promote their growth in vitro. Zygote 31(4), 305-315.
| Crossref | Google Scholar | PubMed |

Nasser LF, Sá Filho MF, Reis EL, Rezende CR, Mapletoft RJ, Bó GA, Baruselli PS (2011) Exogenous progesterone enhances ova and embryo quality following superstimulation of the first follicular wave in Nelore (Bos indicus) donors. Theriogenology 76(2), 320-327.
| Crossref | Google Scholar |

Nivet A-L, Bunel A, Labrecque R, Belanger J, Vigneault C, Blondin P, Sirard M-A (2012) FSH withdrawal improves developmental competence of oocytes in the bovine model. Reproduction 143(2), 165-171.
| Crossref | Google Scholar | PubMed |

O’Doherty AM, O’Shea LC, Fair T (2012) Bovine DNA methylation imprints are established in an oocyte size-specific manner, which are coordinated with the expression of the DNMT3 family proteins. Biology of Reproduction 86, 67.
| Crossref | Google Scholar | PubMed |

O’Doherty AM, O’Gorman A, al Naib A, Brennan L, Daly E, Duffy P, Fair T (2014) Negative energy balance affects imprint stability in oocytes recovered from postpartum dairy cows. Genomics 104, 177-185.
| Crossref | Google Scholar | PubMed |

O’Doherty AM, McGettigan P, Irwin RE, Magee DA, Gagne D, Fournier E, Al-Naib A, Sirard M-A, Walsh CP, Robert C, Fair T (2018) Intragenic sequences in the trophectoderm harbour the greatest proportion of methylation errors in day 17 bovine conceptuses generated using assisted reproductive technologies. BMC Genomics 19(1), 438.
| Crossref | Google Scholar | PubMed |

O’Shea LC, Hensey C, Fair T (2013) Progesterone regulation of AVEN protects bovine oocytes from apoptosis during meiotic maturation. Biology of Reproduction 89(6), 146.
| Crossref | Google Scholar | PubMed |

O’Shea LC, Daly E, Hensey C, Fair T (2017) ATRX is a novel progesterone-regulated protein and biomarker of low developmental potential in mammalian oocytes. Reproduction 153(5), 671-682.
| Crossref | Google Scholar | PubMed |

Pangas SA, Rajkovic A (2006) Transcriptional regulation of early oogenesis: in search of masters. Human Reproduction Update 12(1), 65-76.
| Crossref | Google Scholar | PubMed |

Pieterse MC, Kappen KA, Kruip TAM, Taverne MAM (1988) Aspiration of bovine oocytes during transvaginal ultrasound scanning of the ovaries. Theriogenology 30(4), 751-762.
| Crossref | Google Scholar | PubMed |

Pincus G, Enzmann EV (1935) The comparative behavior of mammalian eggs in vivo and in vitro: I. The activation of ovarian eggs. Journal of Experimental Medicine 62(5), 665-675.
| Crossref | Google Scholar | PubMed |

Razza EM, Pedersen HS, Stroebech L, Fontes PK, Kadarmideen HN, Callesen H, Pihl M, Nogueira MFg, Hyttel P (2019) Simulated physiological oocyte maturation has side effects on bovine oocytes and embryos. Journal of Assisted Reproduction and Genetics 36, 413-424.
| Crossref | Google Scholar | PubMed |

Reddy P, Liu L, Adhikari D, Jagarlamudi K, Rajareddy S, Shen Y, Du C, Tang W, Hämäläinen T, Peng SL, Lan Z-J, Cooney AJ, Huhtaniemi I, Liu K (2008) Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science 319(5863), 611-613.
| Crossref | Google Scholar | PubMed |

Richani D, Gilchrist RB (2018) The epidermal growth factor network: role in oocyte growth, maturation and developmental competence. Human Reproduction Update 24(1), 1-14.
| Crossref | Google Scholar | PubMed |

Rivera FA, Mendonça LG, Lopes G, Jr, Santos JE, Perez RV, Amstalden M, et al. (2011) Reduced progesterone concentration during growth of the first follicular wave affects embryo quality but has no effect on embryo survival post transfer in lactating dairy cows. Reproduction 141(3), 333-342.
| Crossref | Google Scholar |

Rizos D, Ward F, Duffy P, Boland MP, Lonergan P (2002) Consequences of bovine oocyte maturation, fertilization or early embryo development in vitro versus in vivo: implications for blastocyst yield and blastocyst quality. Molecular Reproduction & Development 61(2), 234-248.
| Crossref | Google Scholar | PubMed |

Rose UM, Hanssen RGJM, Kloosterboer HJ (1999) Development and characterization of an in vitro ovulation model using mouse ovarian follicles. Biology of Reproduction 61(2), 503-511.
| Crossref | Google Scholar | PubMed |

Saitou M, Hayashi K (2021) Mammalian in vitro gametogenesis. Science 374(6563), eaaz6830.
| Crossref | Google Scholar |

Sales JNS, Iguma LT, Batista RITP, Quintão CCR, Gama MAS, Freitas C, Pereira MM, Camargo LSA, Viana JHM, Souza JC, Baruselli PS (2015) Effects of a high-energy diet on oocyte quality and in vitro embryo production in Bos indicus and Bos taurus cows. Journal of Dairy Science 98(5), 3086-3099.
| Crossref | Google Scholar | PubMed |

Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, Ohta H, Moritoki Y, Iwatani C, Tsuchiya H, Nakamura S, Sekiguchi K, Sakuma T, Yamamoto T, Mori T, Woltjen K, Nakagawa M, Yamamoto T, Takahashi K, Yamanaka S, Saitou M (2015) Robust in vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell 17(2), 178-194.
| Crossref | Google Scholar | PubMed |

Savio JD, Thatcher WW, Badinga L, de la Sota RL, Wolfenson D (1993) Regulation of dominant follicle turnover during the oestrous cycle in cows. Reproduction 97(1), 197-203.
| Crossref | Google Scholar |

Schams D, Karg H (1969) Radioimmunologische LH-bestimmung im blutserum vom rind unter besonderer berücksichtigung des brunstzyklus. Acta Endocrinologica 61(1), 96-103.
| Crossref | Google Scholar | PubMed |

Simmons R, Tutt DAR, Guven-Ates G, Kwong WY, Labrecque R, Randi F, Sinclair KD (2023) Enhanced progesterone support during stimulated cycles of transvaginal follicular aspiration improves bovine in vitro embryo production. Theriogenology 199, 77-85.
| Crossref | Google Scholar | PubMed |

Sirard MA, Blondin P (1996) Oocyte maturation and IVF in cattle. Animal Reproduction Science 42(1-4), 417-426.
| Crossref | Google Scholar |

Sirois J, Fortune JE (1990) Lengthening the bovine estrous cycle with low levels of exogenous progesterone: a model for studying ovarian follicular dominance. Endocrinology 127(2), 916-925.
| Crossref | Google Scholar | PubMed |

So C, Seres KB, Steyer AM, Mönnich E, Clift D, Pejkovska A, Möbius W, Schuh M (2019) A liquid-like spindle domain promotes acentrosomal spindle assembly in mammalian oocytes. Science 364(6447), eaat9557.
| Crossref | Google Scholar |

Spears N, Boland NI, Murray AA, Gosden RG (1994) Mouse oocytes derived from in vitro grown primary ovarian follicles are fertile. Human Reproduction 9, 527-532.
| Crossref | Google Scholar | PubMed |

Stock AE, Fortune JE (1993) Ovarian follicular dominance in cattle: relationship between prolonged growth of the ovulatory follicle and endocrine parameters. Endocrinology 132, 1108-1114.
| Crossref | Google Scholar | PubMed |

Strange A, Alberio R (2023) Review: A barnyard in the lab: prospect of generating animal germ cells for breeding and conservation. Animal 17(Suppl 1), 100753.
| Crossref | Google Scholar | PubMed |

Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, III, Hao Y, Stoeckius M, Smibert P, Satija R (2019) Comprehensive integration of single-cell data. Cell 177(7), 1888-1902.e21.
| Crossref | Google Scholar | PubMed |

Sun Q-Y, Schatten H (2006) Regulation of dynamic events by microfilaments during oocyte maturation and fertilization. Reproduction 131(2), 193-205.
| Crossref | Google Scholar | PubMed |

Sunderland SJ, Crowe MA, Boland MP, Roche JF, Ireland JJ (1994) Selection, dominance and atresia of follicles during the oestrous cycle of heifers. Journal of Reproduction and Fertility 101(3), 547-555.
| Crossref | Google Scholar | PubMed |

Sutton-McDowall ML, Gilchrist RB, Thompson JG (2010) The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction 139(4), 685-695.
| Crossref | Google Scholar | PubMed |

Svoboda P, Franke V, Schultz RM (2015) Sculpting the transcriptome during the oocyte-to-embryo transition in mouse. Current Topics in Developmental Biology 113, 305-349.
| Crossref | Google Scholar | PubMed |

Telfer EE, Sakaguchi K, Clarkson YL, McLaughlin M (2020) In vitro growth of immature bovine follicles and oocytes. Reproduction, Fertility and Development 32(2), 1-6.
| Crossref | Google Scholar |

Thibier M, Wagner H-G (2002) World statistics for artificial insemination in cattle. Livestock Production Science 74(2), 203-212.
| Crossref | Google Scholar |

Tríbulo A, Rogan D, Tribulo H, Tribulo R, Alasino RV, Beltramo D, Bianco I, Mapletoft RJ, Bó GA (2011) Superstimulation of ovarian follicular development in beef cattle with a single intramuscular injection of Folltropin-V. Animal Reproduction Science 129(1-2), 7-13.
| Crossref | Google Scholar | PubMed |

Uzbekova S, Arlot-Bonnemains Y, Dupont J, Dalbiès-Tran R, Papillier P, Pennetier S, Thélie A, Perreau C, Mermillod P, Prigent C, Uzbekov R (2008) Spatio-temporal expression patterns of aurora kinases a, b, and c and cytoplasmic polyadenylation-element-binding protein in bovine oocytes during meiotic maturation. Biology of Reproduction 78(2), 218-233.
| Crossref | Google Scholar | PubMed |

Uzbekova S, Almiñana C, Labas V, Teixeira-Gomes A-P, Combes-Soia L, Tsikis G, Carvalho AV, Uzbekov R, Singina G (2020) Protein cargo of extracellular vesicles from bovine follicular fluid and analysis of their origin from different ovarian cells. Frontiers in Veterinary Science 7, 584948.
| Crossref | Google Scholar | PubMed |

Van Blerkom J, Bell H, Weipz D (1990) Cellular and developmental biological aspects of bovine meiotic maturation, fertilization, and preimplantation embryogenesis in vitro. Journal of Electron Microscopy Technique 16(4), 298-323.
| Crossref | Google Scholar | PubMed |

van den Hurk R, Spek ER, Hage WJ, Fair T, Ralph JH, Schotanus K (1998) Ultrastructure and viability of isolated bovine preantral follicles. Human Reproduction Update 4(6), 833-841.
| Crossref | Google Scholar | PubMed |

Veselovska L, Smallwood SA, Saadeh H, Stewart KR, Krueger F, Maupetit-Méhouas S, Arnaud P, Tomizawa S-I, Andrews S, Kelsey G (2015) Deep sequencing and de novo assembly of the mouse oocyte transcriptome define the contribution of transcription to the DNA methylation landscape. Genome Biology 16, 209.
| Crossref | Google Scholar | PubMed |

Viana JHM, Siqueira LGB, Palhao MP, Camargo LSA (2018) Features and perspectives of the brazilian in vitro embryo industry. Animal Reproduction 9(1), 12-18.
| Google Scholar |

Walters AH, Bailey TL, Pearson RE, Gwazdauskas FC (2002) Parity-related changes in bovine follicle and oocyte populations, oocyte quality, and hormones to 90 days postpartum. Journal of Dairy Science 85(4), 824-832.
| Crossref | Google Scholar | PubMed |

Wandji S-A, Eppig JJ, Fortune JE (1996) FSH and growth factors affect the growth and endocrine function in vitro of granulosa cells of bovine preantral follicles. Theriogenology 45(4), 817-832.
| Crossref | Google Scholar | PubMed |

Wang X, Catt S, Pangestu M, Temple-Smith P (2011) Successful in vitro culture of pre-antral follicles derived from vitrified murine ovarian tissue: oocyte maturation, fertilization, and live births. Reproduction 141(2), 183-191.
| Crossref | Google Scholar | PubMed |

Wang Z, Liu C-Y, Zhao Y, Dean J (2020) FIGLA, LHX8 and SOHLH1 transcription factor networks regulate mouse oocyte growth and differentiation. Nucleic Acids Research 48(7), 3525-3541.
| Crossref | Google Scholar | PubMed |

Watanabe YF, de Souza AH, Mingoti RD, Ferreira RM, Batista EOS, Dayan A, Watanabe O, Meirelles FV, Nogueira MFG, Ferraz JBS, Baruselli PS (2017) Number of oocytes retrieved per donor during OPU and its relationship with in vitro embryo production and field fertility following embryo transfer. Animal Reproduction 14, 635-644.
| Crossref | Google Scholar |

Wiltbank MC, Souza AH, Carvalho PD, Cunha AP, Giordano JO, Fricke PM, Baez GM, Diskin MG (2014) Physiological and practical effects of progesterone on reproduction in dairy cattle. Animal 8(Suppl 1), 70-81.
| Crossref | Google Scholar | PubMed |

Yamashiro C, Sasaki K, Yabuta Y, Kojima Y, Nakamura T, Okamoto I, Yokobayashi S, Murase Y, Ishikura Y, Shirane K, Sasaki H, Yamamoto T, Saitou M (2018) Generation of human oogonia from induced pluripotent stem cells in vitro. Science 362(6412), 356-360.
| Crossref | Google Scholar | PubMed |

Yan R, Gu C, You D, Huang Z, Qian J, Yang Q, Cheng X, Zhang L, Wang H, Wang P, Guo F (2021) Decoding dynamic epigenetic landscapes in human oocytes using single-cell multi-omics sequencing. Cell Stem Cell 28(9), 1641-1656.e7.
| Crossref | Google Scholar | PubMed |

Yoshino T, Suzuki T, Nagamatsu G, Yabukami H, Ikegaya M, Kishima M, Kita H, Imamura T, Nakashima K, Nishinakamura R, Tachibana M, Inoue M, Shima Y, Morohashi K-I, Hayashi K (2021) Generation of ovarian follicles from mouse pluripotent stem cells. Science 373(6552), eabe0237.
| Crossref | Google Scholar |

Yu L, Wei Y, Sun H-X, Mahdi AK, Pinzon Arteaga CA, Sakurai M, Schmitz DA, Zheng C, Ballard ED, Li J, Tanaka N, Kohara A, Okamura D, Mutto AA, Gu Y, Ross PJ, Wu J (2021) Derivation of intermediate pluripotent stem cells amenable to primordial germ cell specification. Cell Stem Cell 28(3), 550-567.e12.
| Crossref | Google Scholar | PubMed |

Zheng P, Dean J (2007) Oocyte-specific genes affect folliculogenesis, fertilization, and early development. Seminars in Reproductive Medicine 25(4), 243-251.
| Crossref | Google Scholar | PubMed |

Zhu K, Yan L, Zhang X, Lu X, Wang T, Yan J, Liu X, Qiao J, Li L (2015) Identification of a human subcortical maternal complex. Molecular Human Reproduction 21(4), 320-329.
| Crossref | Google Scholar | PubMed |

Zhu Q, Sang F, Withey S, Tang W, Dietmann S, Klisch D, et al. (2021) Specification and epigenomic resetting of the pig germline exhibit conservation with the human lineage. Cell Reports 34(6), 108735.
| Crossref | Google Scholar |