Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW (Open Access)

The ovarian follicle of ruminants: the path from conceptus to adult

Jennifer L. Juengel https://orcid.org/0000-0002-2717-7311 A J , Robert A. Cushman B , Joëlle Dupont C , Stéphane Fabre https://orcid.org/0000-0001-7350-9500 D , Richard G. Lea https://orcid.org/0000-0002-6793-3601 E , Graeme B. Martin F , Francesca Mossa G , Janet L. Pitman H , Christopher A. Price I and Peter Smith A
+ Author Affiliations
- Author Affiliations

A AgResearch Ltd, Invermay Agricultural Centre, Mosgiel, New Zealand.

B Livestock Biosystems Research Unit, US Department of Agriculture, Agricultural Research Service, US Meat Animal Research Center, Clay Center, NE, USA.*

C INRAE Institute UMR85 Physiologie de la Reproduction et des Comportements, Tours University, France.

D GenPhySE, Université de Toulouse, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement, Institut national polytechnique de Toulouse, Ecole nationale vétérinaire de Toulouse, Castanet Tolosan, France.

E School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK.

F UWA Institute of Agriculture, University of Western Australia, Perth, WA, Australia.

G Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Italy.

H School of Biological Sciences, Victoria University of Wellington, New Zealand.

I Faculty of Veterinary Medicine, Université de Montréal, Montréal, QC, Canada.

J Corresponding author.# Email: jenny.juengel@agresearch.co.nz

Reproduction, Fertility and Development 33(10) 621-642 https://doi.org/10.1071/RD21086
Submitted: 22 March 2021  Accepted: 6 June 2021   Published: 2 July 2021

Journal Compilation © CSIRO 2021 Open Access CC BY-NC-ND

Abstract

This review resulted from an international workshop and presents a consensus view of critical advances over the past decade in our understanding of follicle function in ruminants. The major concepts covered include: (1) the value of major genes; (2) the dynamics of fetal ovarian development and its sensitivity to nutritional and environmental influences; (3) the concept of an ovarian follicle reserve, aligned with the rise of anti-Müllerian hormone as a controller of ovarian processes; (4) renewed recognition of the diverse and important roles of theca cells; (5) the importance of follicular fluid as a microenvironment that determines oocyte quality; (6) the ‘adipokinome’ as a key concept linking metabolic inputs with follicle development; and (7) the contribution of follicle development to the success of conception. These concepts are important because, in sheep and cattle, ovulation rate is tightly regulated and, as the primary determinant of litter size, it is a major component of reproductive efficiency and therefore productivity. Nowadays, reproductive efficiency is also a target for improving the ‘methane efficiency’ of livestock enterprises, increasing the need to understand the processes of ovarian development and folliculogenesis, while avoiding detrimental trade-offs as greater performance is sought.

Keywords: fetal programming, genetics, granulosa cell, nutrition, oocyte, theca cell.


References

Aad, P. Y., Echternkamp, S. E., and Spicer, L. J. (2013). Possible role of IGF2 receptors in regulating selection of 2 dominant follicles in cattle selected for twin ovulations and births. Domest. Anim. Endocrinol. 45, 187–195.
Possible role of IGF2 receptors in regulating selection of 2 dominant follicles in cattle selected for twin ovulations and births.Crossref | GoogleScholarGoogle Scholar | 24209503PubMed |

Allan, M. F., Kuehn, L. A., Cushman, R. A., Snelling, W. M., Echternkamp, S. E., and Thallman, R. M. (2009). Confirmation of quantitative trait loci using a low-density single nucleotide polymorphism map for twinning and ovulation rate on bovine chromosome 5. J. Anim. Sci. 87, 46–56.
Confirmation of quantitative trait loci using a low-density single nucleotide polymorphism map for twinning and ovulation rate on bovine chromosome 5.Crossref | GoogleScholarGoogle Scholar | 18791147PubMed |

Amundson, O. L., Fountain, T. H., Larimore, E. L., Richardson, B. N., McNeel, A. K., Wright, E. C., Keisler, D. H., Cushman, R. A., Perry, G. A., and Freetly, H. C. (2015). Postweaning nutritional programming of ovarian development in beef heifers. J. Anim. Sci. 93, 5232–5239.
Postweaning nutritional programming of ovarian development in beef heifers.Crossref | GoogleScholarGoogle Scholar | 26641043PubMed |

Antenos, M., Stemler, M., Boime, I., and Woodruff, T. K. (2007). N-linked oligosaccharides direct the differential assembly and secretion of inhibin alpha- and betaA-subunit dimers. Mol. Endocrinol. 21, 1670–1684.
N-linked oligosaccharides direct the differential assembly and secretion of inhibin alpha- and betaA-subunit dimers.Crossref | GoogleScholarGoogle Scholar | 17456790PubMed |

Anway, M. D., Cupp, A. S., Uzumcu, M., and Skinner, M. K. (2005). Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469.
Epigenetic transgenerational actions of endocrine disruptors and male fertility.Crossref | GoogleScholarGoogle Scholar | 15933200PubMed |

Armstrong, D. T. (1967). On the site of action of luteinizing hormone. Nature 213, 633–634.
On the site of action of luteinizing hormone.Crossref | GoogleScholarGoogle Scholar | 6032265PubMed |

Armstrong, D. T., and Hansel, W. (1959). Alteration of the bovine estrous cycle with oxytocin. J. Dairy Sci. 42, 533–542.
Alteration of the bovine estrous cycle with oxytocin.Crossref | GoogleScholarGoogle Scholar |

Armstrong, D. T., and Grinwich, D. L. (1972). Blockade of spontaneous and LH-induced ovulation in rats by indomethacin, an inhibitor of prostaglandin biosynthesis. Prostaglandins 1, 21–28.
Blockade of spontaneous and LH-induced ovulation in rats by indomethacin, an inhibitor of prostaglandin biosynthesis.Crossref | GoogleScholarGoogle Scholar | 4633871PubMed |

Armstrong, D. T., and Papkoff, H. (1976). Stimulation of aromatization of exogenous and endogenous androgens in ovaries of hypophysectomized rats in vivo by follicle-stimulating hormone. Endocrinology 99, 1144–1151.
Stimulation of aromatization of exogenous and endogenous androgens in ovaries of hypophysectomized rats in vivo by follicle-stimulating hormone.Crossref | GoogleScholarGoogle Scholar | 976193PubMed |

Armstrong, D. T., Goff, A. K., and Dorrington, J. H. (1979). Regulation of follicular estrogen biosynthesis. In ‘Ovarian Follicular Development and Function’. (Eds A. R. Midgley and W. A. Sadler) pp. 169–182. (Raven Press: New York)

Árnason, T., and Jónmundsson, J. V. (2008). Multiple trait genetic evaluation of ewe traits in Icelandic sheep. J. Anim. Breed. Genet. 125, 390–396.
Multiple trait genetic evaluation of ewe traits in Icelandic sheep.Crossref | GoogleScholarGoogle Scholar | 19134074PubMed |

Baltar, A. E., Oliveira, M. A., and Catanho, M. T. (2000). Bovine cumulus/oocyte complex: quantification of LH/hCG receptors. Mol. Reprod. Dev. 55, 433–437.
Bovine cumulus/oocyte complex: quantification of LH/hCG receptors.Crossref | GoogleScholarGoogle Scholar | 10694751PubMed |

Bartlewski, P. M., Baby, T. E., and Giffin, J. L. (2011). Reproductive cycles in sheep. Anim. Reprod. Sci. 124, 259–268.
Reproductive cycles in sheep.Crossref | GoogleScholarGoogle Scholar | 21411253PubMed |

Baruselli, P. S., Batista, E. O. S., Vieira, L. M., and Souza, A. H. (2015). Relationship between follicle population, AMH concentration and fertility in cattle. Anim. Reprod. 12, 487–497.

Bellingham, M., Amezaga, M. R., Mandon-Pepin, B., Speers, C. J., Kyle, C. E., Evans, N. P., Sharpe, R. M., Cotinot, C., Rhind, S. M., and Fowler, P. A. (2013). Exposure to chemical cocktails before or after conception—the effect of timing on ovarian development. Mol. Cell. Endocrinol. 376, 156–172.
Exposure to chemical cocktails before or after conception—the effect of timing on ovarian development.Crossref | GoogleScholarGoogle Scholar | 23791816PubMed |

Bongrani, A., Mellouk, N., Rame, C., Cornuau, M., Guerif, F., Froment, P., and Dupont, J. (2019). Ovarian Expression of Adipokines in Polycystic Ovary Syndrome: A Role for Chemerin, Omentin, and Apelin in Follicular Growth Arrest and Ovulatory Dysfunction? Int. J. Mol. Sci. 20, 3778.
Ovarian Expression of Adipokines in Polycystic Ovary Syndrome: A Role for Chemerin, Omentin, and Apelin in Follicular Growth Arrest and Ovulatory Dysfunction?Crossref | GoogleScholarGoogle Scholar |

Bonnet, A., Bevilacqua, C., Benne, F., Bodin, L., Cotinot, C., Liaubet, L., Sancristobal, M., Sarry, J., Terenina, E., Martin, P., Tosser-Klopp, G., and Mandon-Pepin, B. (2011). Transcriptome profiling of sheep granulosa cells and oocytes during early follicular development obtained by laser capture microdissection. BMC Genomics 12, 417.
Transcriptome profiling of sheep granulosa cells and oocytes during early follicular development obtained by laser capture microdissection.Crossref | GoogleScholarGoogle Scholar | 21851638PubMed |

Borg, R. C., Notter, D. R., and Kott, R. W. (2009). Phenotypic and genetic associations between lamb growth traits and adult ewe body weights in western range sheep. J. Anim. Sci. 87, 3506–3514.
Phenotypic and genetic associations between lamb growth traits and adult ewe body weights in western range sheep.Crossref | GoogleScholarGoogle Scholar | 19648484PubMed |

Borwick, S. C., Rhind, S. M., McMillen, S. R., and Racey, P. A. (1997). Effect of undernutrition of ewes from the time of mating on fetal ovarian development in mid gestation. Reprod. Fertil. Dev. 9, 711–715.
Effect of undernutrition of ewes from the time of mating on fetal ovarian development in mid gestation.Crossref | GoogleScholarGoogle Scholar | 9623491PubMed |

Butler, W. R. (2000). Nutritional interactions with reproductive performance in dairy cattle. Anim. Reprod. Sci. 60–61, 449–457.
Nutritional interactions with reproductive performance in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 10844215PubMed |

Campbell, B. K., Clinton, M., and Webb, R. (2012). The role of anti-Mullerian hormone (AMH) during follicle development in a monovulatory species (sheep). Endocrinology 153, 4533–4543.
The role of anti-Mullerian hormone (AMH) during follicle development in a monovulatory species (sheep).Crossref | GoogleScholarGoogle Scholar | 22778215PubMed |

Campen, K. A., Kucharczyk, K. M., Bogin, B., Ehrlich, J. M., and Combelles, C. M. H. (2018). Spindle abnormalities and chromosome misalignment in bovine oocytes after exposure to low doses of bisphenol A or bisphenol S. Hum. Reprod. 33, 895–904.
Spindle abnormalities and chromosome misalignment in bovine oocytes after exposure to low doses of bisphenol A or bisphenol S.Crossref | GoogleScholarGoogle Scholar | 29538760PubMed |

Cardoso, F. C., Kalscheur, K. F., and Drackley, J. K. (2020). Symposium review: Nutrition strategies for improved health, production, and fertility during the transition period. J. Dairy Sci. 103, 5684–5693.
Symposium review: Nutrition strategies for improved health, production, and fertility during the transition period.Crossref | GoogleScholarGoogle Scholar | 32008772PubMed |

Castilho, A. C. S., Price, C. A., Dalanezi, F., Ereno, R. L., Machado, M. F., Barros, C. M., Gasperin, B. G., Goncalves, P. B. D., and Buratini, J. (2017). Evidence that fibroblast growth factor 10 plays a role in follicle selection in cattle. Reprod. Fertil. Dev. 29, 234–243.
Evidence that fibroblast growth factor 10 plays a role in follicle selection in cattle.Crossref | GoogleScholarGoogle Scholar |

Chagas, L. M., Bass, J. J., Blache, D., Burke, C. R., Kay, J. K., Lindsay, D. R., Lucy, M. C., Martin, G. B., Meier, S., Rhodes, F. M., Roche, J. R., Thatcher, W. W., and Webb, R. (2007). Invited review: New perspectives on the roles of nutrition and metabolic priorities in the subfertility of high-producing dairy cows. J. Dairy Sci. 90, 4022–4032.
Invited review: New perspectives on the roles of nutrition and metabolic priorities in the subfertility of high-producing dairy cows.Crossref | GoogleScholarGoogle Scholar | 17699018PubMed |

Chang, H. M., Qiao, J., and Leung, P. C. (2016a). Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors. Hum. Reprod. Update 23, 1–18.
Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors.Crossref | GoogleScholarGoogle Scholar | 27797914PubMed |

Chang, S. S., Eisenberg, D., Zhao, L., Adams, C., Leib, R., Morser, J., and Leung, L. (2016b). Chemerin activation in human obesity. Obesity 24, 1522–1529.
Chemerin activation in human obesity.Crossref | GoogleScholarGoogle Scholar | 27222113PubMed |

Chantepie, L., Bordes, A., Aletru, M., Burg, C., Debat, F., Rialland, F., Rivemale, F., Tadi, N., Alabart, J. L., Lahoz, B., and Fabre, S. (2018). Search for a pleiotropic effect of the FecLL prolific mutation in Lacaune meat sheep. Reprod. Domest. Anim. 53, 86–87.
Search for a pleiotropic effect of the FecLL prolific mutation in Lacaune meat sheep.Crossref | GoogleScholarGoogle Scholar |

Chianese, R., Troisi, J., Richards, S., Scafuro, M., Fasano, S., Guida, M., Pierantoni, R., and Meccariello, R. (2018). Bisphenol A in reproduction: Epigenetic effects. Curr. Med. Chem. 25, 748–770.
Bisphenol A in reproduction: Epigenetic effects.Crossref | GoogleScholarGoogle Scholar | 28990514PubMed |

Clark, Z. L., Heath, D. A., O’Connell, A. R., Juengel, J. L., McNatty, K. P., and Pitman, J. L. (2020). The follicular microenvironment in low (++) and high (I+B+) ovulation rate ewes. Reproduction 159, 585–599.
The follicular microenvironment in low (++) and high (I+B+) ovulation rate ewes.Crossref | GoogleScholarGoogle Scholar | 32069217PubMed |

Crisosto, N., Sir-Petermann, T., Greiner, M., Maliqueo, M., Moreno, M., Aedo, P., and Lara, H. E. (2009). Testosterone-induced downregulation of anti-Mullerian hormone expression in granulosa cells from small bovine follicles. Endocrine 36, 339–345.
Testosterone-induced downregulation of anti-Mullerian hormone expression in granulosa cells from small bovine follicles.Crossref | GoogleScholarGoogle Scholar | 19714502PubMed |

Cruz, G., Foster, W., Paredes, A., Yi, K. D., and Uzumcu, M. (2014). Long-term effects of early-life exposure to environmental oestrogens on ovarian function: role of epigenetics. J. Neuroendocrinol. 26, 613–624.
Long-term effects of early-life exposure to environmental oestrogens on ovarian function: role of epigenetics.Crossref | GoogleScholarGoogle Scholar | 25040227PubMed |

Cushman, R. A., McNeel, A. K., and Freetly, H. C. (2014). The impact of cow nutrient status during the second and third trimesters on age at puberty, antral follicle count, and fertility of daughters. Livest. Sci. 162, 252–258.
The impact of cow nutrient status during the second and third trimesters on age at puberty, antral follicle count, and fertility of daughters.Crossref | GoogleScholarGoogle Scholar |

D’Occhio, M. J., Baruselli, P. S., and Campanile, G. (2019). Influence of nutrition, body condition, and metabolic status on reproduction in female beef cattle: A review. Theriogenology 125, 277–284.
Influence of nutrition, body condition, and metabolic status on reproduction in female beef cattle: A review.Crossref | GoogleScholarGoogle Scholar | 30497026PubMed |

D’Occhio, M. J., Campanile, G., and Baruselli, P. S. (2020). Transforming growth factor-beta superfamily and interferon-tau in ovarian function and embryo development in female cattle: review of biology and application. Reprod. Fertil. Dev. 32, 539–552.
Transforming growth factor-beta superfamily and interferon-tau in ovarian function and embryo development in female cattle: review of biology and application.Crossref | GoogleScholarGoogle Scholar |

Dai, Y., Ivell, R., and Anand-Ivell, R. (2017). Theca cell INSL3 and steroids together orchestrate the growing bovine antral follicle. Front. Physiol. 8, 1033.
Theca cell INSL3 and steroids together orchestrate the growing bovine antral follicle.Crossref | GoogleScholarGoogle Scholar | 29311967PubMed |

Deniz, R., Yavuzkir, S., Ugur, K., Ustebay, D. U., Baykus, Y., Ustebay, S., and Aydin, S. (2021). Subfatin and asprosin, two new metabolic players of polycystic ovary syndrome. J. Obstet. Gynaecol. 41, 279–284.
Subfatin and asprosin, two new metabolic players of polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 32608281PubMed |

Diskin, M. G., and Morris, D. G. (2008). Embryonic and early foetal losses in cattle and other ruminants. Reprod. Domest. Anim. 43, 260–267.
Embryonic and early foetal losses in cattle and other ruminants.Crossref | GoogleScholarGoogle Scholar | 18638133PubMed |

Drouilhet, L., Taragnat, C., Fontaine, J., Duittoz, A., Mulsant, P., Bodin, L., and Fabre, S. (2010). Endocrine characterization of the reproductive axis in highly prolific lacaune sheep homozygous for the FecLL mutation. Biol. Reprod. 82, 815–824.
Endocrine characterization of the reproductive axis in highly prolific lacaune sheep homozygous for the FecLL mutation.Crossref | GoogleScholarGoogle Scholar | 20075395PubMed |

Drouilhet, L., Mansanet, C., Sarry, J., Tabet, K., Bardou, P., Woloszyn, F., Lluch, J., Harichaux, G., Viguie, C., Monniaux, D., Bodin, L., Mulsant, P., and Fabre, S. (2013). The highly prolific phenotype of Lacaune sheep is associated with an ectopic expression of the B4GALNT2 gene within the ovary. PLoS Genet. 9, e1003809.
The highly prolific phenotype of Lacaune sheep is associated with an ectopic expression of the B4GALNT2 gene within the ovary.Crossref | GoogleScholarGoogle Scholar | 24086150PubMed |

Dupont, J., Scaramuzzi, R. J., and Reverchon, M. (2014). The effect of nutrition and metabolic status on the development of follicles, oocytes and embryos in ruminants. Animal 8, 1031–1044.
The effect of nutrition and metabolic status on the development of follicles, oocytes and embryos in ruminants.Crossref | GoogleScholarGoogle Scholar | 24774511PubMed |

Echternkamp, S. E., Spicer, L. J., Gregory, K. E., Canning, S. F., and Hammond, J. M. (1990). Concentrations of insulin-like growth factor-I in blood and ovarian follicular fluid of cattle selected for twins. Biol. Reprod. 43, 8–14.
Concentrations of insulin-like growth factor-I in blood and ovarian follicular fluid of cattle selected for twins.Crossref | GoogleScholarGoogle Scholar | 2393694PubMed |

Echternkamp, S. E., Roberts, A. J., Lunstra, D. D., Wise, T., and Spicer, L. J. (2004). Ovarian follicular development in cattle selected for twin ovulations and births. J. Anim. Sci. 82, 459–471.
Ovarian follicular development in cattle selected for twin ovulations and births.Crossref | GoogleScholarGoogle Scholar | 14974544PubMed |

Eichenlaub-Ritter, U., and Pacchierotti, F. (2015). Bisphenol A effects on mammalian oogenesis and epigenetic integrity of oocytes: A case study exploring risks of endocrine disrupting chemicals. BioMed Res. Int. 2015, 698795.
Bisphenol A effects on mammalian oogenesis and epigenetic integrity of oocytes: A case study exploring risks of endocrine disrupting chemicals.Crossref | GoogleScholarGoogle Scholar | 26339634PubMed |

Eisler, M. C., Lee, M. R., Tarlton, J. F., Martin, G. B., Beddington, J., Dungait, J. A., Greathead, H., Liu, J., Mathew, S., Miller, H., Misselbrook, T., Murray, P., Vinod, V. K., Van Saun, R., and Winter, M. (2014). Agriculture: Steps to sustainable livestock. Nature 507, 32–34.
Agriculture: Steps to sustainable livestock.Crossref | GoogleScholarGoogle Scholar | 24605375PubMed |

El-Sheikh Ali, H., Kitahara, G., Takahashi, T., Mido, S., Sadawy, M., Kobayashi, I., Hemmi, K., and Osawa, T. (2017). Plasma anti-Mullerian hormone profile in heifers from birth through puberty and relationship with puberty onset. Biol. Reprod. 97, 153–161.
Plasma anti-Mullerian hormone profile in heifers from birth through puberty and relationship with puberty onset.Crossref | GoogleScholarGoogle Scholar | 28859283PubMed |

Eppig, J. J. (1993) Regulation of mammalian oocyte maturation. In ‘The Ovary’. (Eds E. Y. Adashi and P. C. K. Leung) pp. 185–208. (Raven Press Ltd.: New York)

Eppig, J. J., Wigglesworth, K., Pendola, F., and Hirao, Y. (1997). Murine oocytes suppress expression of luteinizing hormone receptor messenger ribonucleic acid by granulosa cells. Biol. Reprod. 56, 976–984.
Murine oocytes suppress expression of luteinizing hormone receptor messenger ribonucleic acid by granulosa cells.Crossref | GoogleScholarGoogle Scholar | 9096881PubMed |

Erickson, B. H. (1966). Development and senescence of the postnatal bovine ovary. J. Anim. Sci. 25, 800–805.
Development and senescence of the postnatal bovine ovary.Crossref | GoogleScholarGoogle Scholar | 6007918PubMed |

Estienne, A., Pierre, A., di Clemente, N., Picard, J. Y., Jarrier, P., Mansanet, C., Monniaux, D., and Fabre, S. (2015). Anti-Mullerian hormone regulation by the bone morphogenetic proteins in the sheep ovary: deciphering a direct regulatory pathway. Endocrinology 156, 301–313.
Anti-Mullerian hormone regulation by the bone morphogenetic proteins in the sheep ovary: deciphering a direct regulatory pathway.Crossref | GoogleScholarGoogle Scholar | 25322464PubMed |

Evans, A. C., Mossa, F., Walsh, S. W., Scheetz, D., Jimenez-Krassel, F., Ireland, J. L., Smith, G. W., and Ireland, J. J. (2012). Effects of maternal environment during gestation on ovarian folliculogenesis and consequences for fertility in bovine offspring. Reprod. Domest. Anim. 47, 31–37.
Effects of maternal environment during gestation on ovarian folliculogenesis and consequences for fertility in bovine offspring.Crossref | GoogleScholarGoogle Scholar | 22827347PubMed |

Fabre, S., Pierre, A., Mulsant, P., Bodin, L., Di Pasquale, E., Persani, L., Monget, P., and Monniaux, D. (2006). Regulation of ovulation rate in mammals: contribution of sheep genetic models. Reprod. Biol. Endocrinol. 4, 20.
Regulation of ovulation rate in mammals: contribution of sheep genetic models.Crossref | GoogleScholarGoogle Scholar | 16611365PubMed |

Ferdousy, R. N., Kereilwe, O., and Kadokawa, H. (2020). Anti-Müllerian hormone is expressed and secreted by bovine oviductal and endometrial epithelial cells. Anim. Sci. J. 91, e13456.
Anti-Müllerian hormone is expressed and secreted by bovine oviductal and endometrial epithelial cells.Crossref | GoogleScholarGoogle Scholar |

Ferris, J., Mahboubi, K., MacLusky, N., King, W. A., and Favetta, L. A. (2016). BPA exposure during in vitro oocyte maturation results in dose-dependent alterations to embryo development rates, apoptosis rate, sex ratio and gene expression. Reprod. Toxicol. 59, 128–138.
BPA exposure during in vitro oocyte maturation results in dose-dependent alterations to embryo development rates, apoptosis rate, sex ratio and gene expression.Crossref | GoogleScholarGoogle Scholar | 26686065PubMed |

Forde, N., Beltman, M. E., Lonergan, P., Diskin, M., Roche, J. F., and Crowe, M. A. (2011). Oestrous cycles in Bos taurus cattle. Anim. Reprod. Sci. 124, 163–169.
Oestrous cycles in Bos taurus cattle.Crossref | GoogleScholarGoogle Scholar | 20875708PubMed |

Fowler, P. A., Dora, N. J., McFerran, H., Amezaga, M. R., Miller, D. W., Lea, R. G., Cash, P., McNeilly, A. S., Evans, N. P., Cotinot, C., Sharpe, R. M., and Rhind, S. M. (2008). In utero exposure to low doses of environmental pollutants disrupts fetal ovarian development in sheep. Mol. Hum. Reprod. 14, 269–280.
In utero exposure to low doses of environmental pollutants disrupts fetal ovarian development in sheep.Crossref | GoogleScholarGoogle Scholar | 18436539PubMed |

Franciosi, F., Coticchio, G., Lodde, V., Tessaro, I., Modina, S. C., Fadini, R., Dal Canto, M., Renzini, M. M., Albertini, D. F., and Luciano, A. M. (2014). Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol. Reprod. 91, 61.
Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes.Crossref | GoogleScholarGoogle Scholar | 25078681PubMed |

Freetly, H. C., Vonnahme, K. A., McNeel, A. K., Camacho, L. E., Amundson, O. L., Forbes, E. D., Lents, C. A., and Cushman, R. A. (2014). The consequence of level of nutrition on heifer ovarian and mammary development. J. Anim. Sci. 92, 5437–5443.
The consequence of level of nutrition on heifer ovarian and mammary development.Crossref | GoogleScholarGoogle Scholar | 25403194PubMed |

Freetly, H. C., Cushman, R. A., and Bennett, G. L. (2021). Production performance of cows raised with different post weaning growth patterns. Transl. Anim. Sci , .
Production performance of cows raised with different post weaning growth patterns.Crossref | GoogleScholarGoogle Scholar | 33659864PubMed |

García-Guerra, A., Motta, J. C. L., Melo, L. F., Kirkpatrick, B. W., and Wiltbank, M. C. (2017). Ovulation rate, antral follicle count, and circulating anti-Mullerian hormone in Trio allele carriers, a novel high fecundity bovine genotype. Theriogenology 101, 81–90.
Ovulation rate, antral follicle count, and circulating anti-Mullerian hormone in Trio allele carriers, a novel high fecundity bovine genotype.Crossref | GoogleScholarGoogle Scholar | 28708520PubMed |

García-Guerra, A., Canavessi, A. M. O., Monteiro, P. L. J., Mezera, M. A., Sartori, R., Kirkpatrick, B. W., and Wiltbank, M. C. (2018a). Trio, a novel bovine high fecundity allele: III. Acquisition of dominance and ovulatory capacity at a smaller follicle size. Biol. Reprod. 98, 350–365.
Trio, a novel bovine high fecundity allele: III. Acquisition of dominance and ovulatory capacity at a smaller follicle size.Crossref | GoogleScholarGoogle Scholar | 29425314PubMed |

García-Guerra, A., Kamalludin, M. H., Kirkpatrick, B. W., and Wiltbank, M. C. (2018b). Trio a novel bovine high-fecundity allele: II. Hormonal profile and follicular dynamics underlying the high ovulation rate. Biol. Reprod. 98, 335–349.
Trio a novel bovine high-fecundity allele: II. Hormonal profile and follicular dynamics underlying the high ovulation rate.Crossref | GoogleScholarGoogle Scholar | 29425274PubMed |

García-Guerra, A., Wiltbank, M. C., Battista, S. E., Kirkpatrick, B. W., and Sartori, R. (2018c). Mechanisms regulating follicle selection in ruminants: lessons learned from multiple ovulation models. Anim. Reprod. 15, 660–679.
Mechanisms regulating follicle selection in ruminants: lessons learned from multiple ovulation models.Crossref | GoogleScholarGoogle Scholar |

Gilchrist, R. B., Lane, M., and Thompson, J. G. (2008). Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum. Reprod. Update 14, 159–177.
Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality.Crossref | GoogleScholarGoogle Scholar | 18175787PubMed |

Gilchrist, R. B., Luciano, A. M., Richani, D., Zeng, H. T., Wang, X., Vos, M. D., Sugimura, S., Smitz, J., Richard, F. J., and Thompson, J. G. (2016). Oocyte maturation and quality: role of cyclic nucleotides. Reproduction 152, R143–R157.
Oocyte maturation and quality: role of cyclic nucleotides.Crossref | GoogleScholarGoogle Scholar | 27422885PubMed |

Glister, C., Kemp, C. F., and Knight, P. G. (2004). Bone morphogenetic protein (BMP) ligands and receptors in bovine ovarian follicle cells: actions of BMP-4, -6 and -7 on granulosa cells and differential modulation of Smad-1 phosphorylation by follistatin. Reproduction 127, 239–254.
Bone morphogenetic protein (BMP) ligands and receptors in bovine ovarian follicle cells: actions of BMP-4, -6 and -7 on granulosa cells and differential modulation of Smad-1 phosphorylation by follistatin.Crossref | GoogleScholarGoogle Scholar | 15056790PubMed |

Glister, C., Richards, S. L., and Knight, P. G. (2005). Bone morphogenetic proteins (BMP) -4, -6, and -7 potently suppress basal and luteinizing hormone-induced androgen production by bovine theca interna cells in primary culture: could ovarian hyperandrogenic dysfunction be caused by a defect in thecal BMP signaling? Endocrinology 146, 1883–1892.
Bone morphogenetic proteins (BMP) -4, -6, and -7 potently suppress basal and luteinizing hormone-induced androgen production by bovine theca interna cells in primary culture: could ovarian hyperandrogenic dysfunction be caused by a defect in thecal BMP signaling?Crossref | GoogleScholarGoogle Scholar | 15625241PubMed |

Glister, C., Satchell, L., and Knight, P. G. (2011). Granulosal and thecal expression of bone morphogenetic protein- and activin-binding protein mRNA transcripts during bovine follicle development and factors modulating their expression in vitro. Reproduction 142, 581–591.
Granulosal and thecal expression of bone morphogenetic protein- and activin-binding protein mRNA transcripts during bovine follicle development and factors modulating their expression in vitro.Crossref | GoogleScholarGoogle Scholar | 21821720PubMed |

Glister, C., Satchell, L., Bathgate, R. A., Wade, J. D., Dai, Y., Ivell, R., Anand-Ivell, R., Rodgers, R. J., and Knight, P. G. (2013). Functional link between bone morphogenetic proteins and insulin-like peptide 3 signaling in modulating ovarian androgen production. Proc. Natl. Acad. Sci. USA 110, E1426–E1435.
Functional link between bone morphogenetic proteins and insulin-like peptide 3 signaling in modulating ovarian androgen production.Crossref | GoogleScholarGoogle Scholar | 23530236PubMed |

Glister, C., Regan, S. L., Samir, M., and Knight, P. (2019). Gremlin, Noggin, Chordin and follistatin differentially modulate BMP induced suppression of androgen secretion by bovine ovarian theca cells. J. Mol. Endocrinol. 62, 15–25.
Gremlin, Noggin, Chordin and follistatin differentially modulate BMP induced suppression of androgen secretion by bovine ovarian theca cells.Crossref | GoogleScholarGoogle Scholar |

Gobikrushanth, M., Purfield, D. C., Colazo, M. G., Butler, S. T., Wang, Z., and Ambrose, D. J. (2018). The relationship between serum anti-Mullerian hormone concentrations and fertility, and genome-wide associations for anti-Mullerian hormone in Holstein cows. J. Dairy Sci. 101, 7563–7574.
The relationship between serum anti-Mullerian hormone concentrations and fertility, and genome-wide associations for anti-Mullerian hormone in Holstein cows.Crossref | GoogleScholarGoogle Scholar | 29729909PubMed |

Gobikrushanth, M., Purfield, D. C., Canadas, E. R., Herlihy, M. M., Kenneally, J., Murray, M., Kearney, F. J., Colazo, M. G., Ambrose, D. J., and Butler, S. T. (2019). Anti-Mullerian hormone in grazing dairy cows: Identification of factors affecting plasma concentration, relationship with phenotypic fertility, and genome-wide associations. J. Dairy Sci. 102, 11622–11635.
Anti-Mullerian hormone in grazing dairy cows: Identification of factors affecting plasma concentration, relationship with phenotypic fertility, and genome-wide associations.Crossref | GoogleScholarGoogle Scholar | 31521342PubMed |

Grynberg, M., Pierre, A., Rey, R., Leclerc, A., Arouche, N., Hesters, L., Catteau-Jonard, S., Frydman, R., Picard, J. Y., Fanchin, R., Veitia, R., di Clemente, N., and Taieb, J. (2012). Differential regulation of ovarian anti-mullerian hormone (AMH) by estradiol through alpha- and beta-estrogen receptors. J. Clin. Endocrinol. Metab. 97, E1649–E1657.
Differential regulation of ovarian anti-mullerian hormone (AMH) by estradiol through alpha- and beta-estrogen receptors.Crossref | GoogleScholarGoogle Scholar | 22689696PubMed |

Guerrero-Netro, H. M., Estienne, A., Chorfi, Y., and Price, C. A. (2017). The mycotoxin metabolite deepoxy- deoxynivalenol increases apoptosis and decreases steroidogenesis in bovine ovarian theca cells. Biol. Reprod. 97, 746–757.
The mycotoxin metabolite deepoxy- deoxynivalenol increases apoptosis and decreases steroidogenesis in bovine ovarian theca cells.Crossref | GoogleScholarGoogle Scholar | 29045588PubMed |

Han, S. J., and Conti, M. (2006). New pathways from PKA to the Cdc2/cyclin B complex in oocytes: Wee1B as a potential PKA substrate. Cell Cycle 5, 227–231.
New pathways from PKA to the Cdc2/cyclin B complex in oocytes: Wee1B as a potential PKA substrate.Crossref | GoogleScholarGoogle Scholar | 16418576PubMed |

Han, P., Guerrero-Netro, H., Estienne, A., and Price, C. A. (2018). Effects of fibroblast growth factors and the transcription factor, early growth response 1, on bovine theca cells. Mol. Cell. Endocrinol. 476, 96–102.
Effects of fibroblast growth factors and the transcription factor, early growth response 1, on bovine theca cells.Crossref | GoogleScholarGoogle Scholar | 29723542PubMed |

Hatzirodos, N., Irving-Rodgers, H. F., Hummitzsch, K., and Rodgers, R. J. (2014). Transcriptome profiling of the theca interna from bovine ovarian follicles during atresia. PLoS One 9, e99706.
Transcriptome profiling of the theca interna from bovine ovarian follicles during atresia.Crossref | GoogleScholarGoogle Scholar | 24956388PubMed |

Hernandez-Medrano, J. H., Campbell, B. K., and Webb, R. (2012). Nutritional influences on folliculogenesis. Reprod. Domest. Anim. 47, 274–282.
Nutritional influences on folliculogenesis.Crossref | GoogleScholarGoogle Scholar | 22827381PubMed |

Hiradate, Y., Hoshino, Y., Tanemura, K., and Sato, E. (2014). C-type natriuretic peptide inhibits porcine oocyte meiotic resumption. Zygote 22, 372–377.
C-type natriuretic peptide inhibits porcine oocyte meiotic resumption.Crossref | GoogleScholarGoogle Scholar | 23331536PubMed |

Huang, J., and Zeng, H. (2021). The influence of environmental factors on ovarian function, follicular genesis, and oocyte quality. Adv. Exp. Med. Biol. 1300, 41–62.
The influence of environmental factors on ovarian function, follicular genesis, and oocyte quality.Crossref | GoogleScholarGoogle Scholar | 33523429PubMed |

Hussein, T. S., Thompson, J. G., and Gilchrist, R. B. (2006). Oocyte-secreted factors enhance oocyte developmental competence. Dev. Biol. 296, 514–521.
Oocyte-secreted factors enhance oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 16854407PubMed |

(2018). Recipient of the 2018 IETS Pioneer Award: David Thomas Armstrong, BSA, MSc, PhD. Reprod. Fertil. Dev. 30, xxii–xxiv.
Recipient of the 2018 IETS Pioneer Award: David Thomas Armstrong, BSA, MSc, PhD.Crossref | GoogleScholarGoogle Scholar |

Ireland, J. L., Scheetz, D., Jimenez-Krassel, F., Themmen, A. P., Ward, F., Lonergan, P., Smith, G. W., Perez, G. I., Evans, A. C., and Ireland, J. J. (2008). Antral follicle count reliably predicts number of morphologically healthy oocytes and follicles in ovaries of young adult cattle. Biol. Reprod. 79, 1219–1225.
Antral follicle count reliably predicts number of morphologically healthy oocytes and follicles in ovaries of young adult cattle.Crossref | GoogleScholarGoogle Scholar | 18768912PubMed |

Irving-Rodgers, H. F., van Wezel, I. L., Mussard, M. L., Kinder, J. E., and Rodgers, R. J. (2001). Atresia revisited: two basic patterns of atresia of bovine antral follicles. Reproduction 122, 761–775.
Atresia revisited: two basic patterns of atresia of bovine antral follicles.Crossref | GoogleScholarGoogle Scholar | 11690537PubMed |

Jaton, C., Koeck, A., Sargolzaei, M., Malchiodi, F., Price, C. A., Schenkel, F. S., and Miglior, F. (2016). Genetic analysis of superovulatory response of Holstein cows in Canada. J. Dairy Sci. 99, 3612–3623.
Genetic analysis of superovulatory response of Holstein cows in Canada.Crossref | GoogleScholarGoogle Scholar | 26923051PubMed |

Jiang, Z., and Price, C. A. (2012). Differential actions of fibroblast growth factors on intracellular pathways and target gene expression in bovine ovarian granulosa cells. Reproduction 144, 625–632.
Differential actions of fibroblast growth factors on intracellular pathways and target gene expression in bovine ovarian granulosa cells.Crossref | GoogleScholarGoogle Scholar | 22956519PubMed |

Jiang, Z., Guerrero-Netro, H. M., Juengel, J. L., and Price, C. A. (2013). Divergence of intracellular signaling pathways and early response genes of two closely related fibroblast growth factors, FGF8 and FGF18, in bovine ovarian granulosa cells. Mol. Cell. Endocrinol. 375, 97–105.
Divergence of intracellular signaling pathways and early response genes of two closely related fibroblast growth factors, FGF8 and FGF18, in bovine ovarian granulosa cells.Crossref | GoogleScholarGoogle Scholar | 23707615PubMed |

Jimenez-Krassel, F., Folger, J. K., Ireland, J. L., Smith, G. W., Hou, X., Davis, J. S., Lonergan, P., Evans, A. C., and Ireland, J. J. (2009). Evidence that high variation in ovarian reserves of healthy young adults has a negative impact on the corpus luteum and endometrium during estrous cycles in cattle. Biol. Reprod. 80, 1272–1281.
Evidence that high variation in ovarian reserves of healthy young adults has a negative impact on the corpus luteum and endometrium during estrous cycles in cattle.Crossref | GoogleScholarGoogle Scholar | 19211804PubMed |

Juengel, J. L. (2018). How the quest to improve sheep reproduction provided insight into oocyte control of folliculr development. J. R. Soc. N. Z. 48, 143–163.
How the quest to improve sheep reproduction provided insight into oocyte control of folliculr development.Crossref | GoogleScholarGoogle Scholar |

Juengel, J. L., Davis, G. H., and McNatty, K. P. (2013). Using sheep lines with mutations in single genes to better understand ovarian function. Reproduction 146, R111–R123.
Using sheep lines with mutations in single genes to better understand ovarian function.Crossref | GoogleScholarGoogle Scholar | 23801782PubMed |

Juengel, J. L., Smith, P. R., Quirke, L. D., French, M. C., and Edwards, S. J. (2018). The local regulation of folliculogenesis by members of the transforming growth factor superfamily and its relevance for advanced breeding programmes. Anim. Reprod. 15, 180–190.
The local regulation of folliculogenesis by members of the transforming growth factor superfamily and its relevance for advanced breeding programmes.Crossref | GoogleScholarGoogle Scholar | 34178140PubMed |

Kafi, M., Tamadon, A., and Saeb, M. (2015). The relationship between serum adiponectin and postpartum luteal activity in high-producing dairy cows. Theriogenology 83, 1264–1271.
The relationship between serum adiponectin and postpartum luteal activity in high-producing dairy cows.Crossref | GoogleScholarGoogle Scholar | 25680575PubMed |

Kurowska, P., Mlyczyńska, E., Dawid, M., Sierpowski, M., Estienne, A., Dupont, J., and Rak, A. (2021). Adipokines change the balance of proliferation/apoptosis in the ovarian cells of human and domestic animals: a comparative review. Anim. Reprod. Sci. 228, 106737.
Adipokines change the balance of proliferation/apoptosis in the ovarian cells of human and domestic animals: a comparative review.Crossref | GoogleScholarGoogle Scholar | 33756403PubMed |

Lagaly, D. V., Aad, P. Y., Grado-Ahuir, J. A., Hulsey, L. B., and Spicer, L. J. (2008). Role of adiponectin in regulating ovarian theca and granulosa cell function. Mol. Cell. Endocrinol. 284, 38–45.
Role of adiponectin in regulating ovarian theca and granulosa cell function.Crossref | GoogleScholarGoogle Scholar | 18289773PubMed |

Lahoz, B., Alabart, J. L., Jurado, J. J., Calvo, J. H., Martinez-Royo, A., Fantova, E., and Folch, J. (2011). Effect of the FecX(R) polymorphism in the bone morphogenetic protein 15 gene on natural or equine chorionic gonadotropin-induced ovulation rate and litter size in Rasa Aragonesa ewes and implications for on-farm application. J. Anim. Sci. 89, 3522–3530.
Effect of the FecX(R) polymorphism in the bone morphogenetic protein 15 gene on natural or equine chorionic gonadotropin-induced ovulation rate and litter size in Rasa Aragonesa ewes and implications for on-farm application.Crossref | GoogleScholarGoogle Scholar | 21622876PubMed |

Lahoz, B., Alabart, J. L., Monniaux, D., Mermillod, P., and Folch, J. (2012). Anti-Mullerian hormone plasma concentration in prepubertal ewe lambs as a predictor of their fertility at a young age. BMC Vet. Res. 8, 118.
Anti-Mullerian hormone plasma concentration in prepubertal ewe lambs as a predictor of their fertility at a young age.Crossref | GoogleScholarGoogle Scholar | 22824005PubMed |

Lahoz, B., Alabart, J. L., Folch, J., Sanchez, P., Echegoyen, E., and Cocero, M. J. (2013). Influence of the FecX(R) allele in heterozygous ewes on follicular population and outcomes of IVP and ET using LOPU-derived oocytes. Reprod. Domest. Anim. 48, 717–723.
Influence of the FecX(R) allele in heterozygous ewes on follicular population and outcomes of IVP and ET using LOPU-derived oocytes.Crossref | GoogleScholarGoogle Scholar | 23438026PubMed |

Lahoz, B., Alabart, J. L., Cocero, M. J., Monniaux, D., Echegoyen, E., Sanchez, P., and Folch, J. (2014). Anti-Mullerian hormone concentration in sheep and its dependence of age and independence of BMP15 genotype: an endocrine predictor to select the best donors for embryo biotechnologies. Theriogenology 81, 347–357.
Anti-Mullerian hormone concentration in sheep and its dependence of age and independence of BMP15 genotype: an endocrine predictor to select the best donors for embryo biotechnologies.Crossref | GoogleScholarGoogle Scholar | 24268018PubMed |

Lea, R. G., Andrade, L. P., Rae, M. T., Hannah, L. T., Kyle, C. E., Murray, J. F., Rhind, S. M., and Miller, D. W. (2006). Effects of maternal undernutrition during early pregnancy on apoptosis regulators in the ovine fetal ovary. Reproduction 131, 113–124.
Effects of maternal undernutrition during early pregnancy on apoptosis regulators in the ovine fetal ovary.Crossref | GoogleScholarGoogle Scholar | 16388015PubMed |

Lea, R. G., Amezaga, M. R., Loup, B., Mandon-Pepin, B., Stefansdottir, A., Filis, P., Kyle, C., Zhang, Z., Allen, C., Purdie, L., Jouneau, L., Cotinot, C., Rhind, S. M., Sinclair, K. D., and Fowler, P. A. (2016). The fetal ovary exhibits temporal sensitivity to a ‘real-life’ mixture of environmental chemicals. Sci. Rep. 6, 22279.
The fetal ovary exhibits temporal sensitivity to a ‘real-life’ mixture of environmental chemicals.Crossref | GoogleScholarGoogle Scholar | 26931299PubMed |

Maillard, V., Uzbekova, S., Guignot, F., Perreau, C., Rame, C., Coyral-Castel, S., and Dupont, J. (2010). Effect of adiponectin on bovine granulosa cell steroidogenesis, oocyte maturation and embryo development. Reprod. Biol. Endocrinol. 8, 23.
Effect of adiponectin on bovine granulosa cell steroidogenesis, oocyte maturation and embryo development.Crossref | GoogleScholarGoogle Scholar | 20219117PubMed |

Maillard, V., Froment, P., Rame, C., Uzbekova, S., Elis, S., and Dupont, J. (2011). Expression and effect of resistin on bovine and rat granulosa cell steroidogenesis and proliferation. Reproduction 141, 467–479.
Expression and effect of resistin on bovine and rat granulosa cell steroidogenesis and proliferation.Crossref | GoogleScholarGoogle Scholar | 21239528PubMed |

Manikkam, M., Haque, M. M., Guerrero-Bosagna, C., Nilsson, E. E., and Skinner, M. K. (2014). Pesticide methoxychlor promotes the epigenetic transgenerational inheritance of adult-onset disease through the female germline. PLoS One 9, e102091.
Pesticide methoxychlor promotes the epigenetic transgenerational inheritance of adult-onset disease through the female germline.Crossref | GoogleScholarGoogle Scholar | 25057798PubMed |

Martin, G. B., Scaramuzzi, R. J., and Henstridge, J. D. (1983). Effects of oestradiol, progesterone and androstenedione on the pulsatile secretion of luteinizing hormone in ovariectomized ewes during spring and autumn. J. Endocrinol. 96, 181–193.
Effects of oestradiol, progesterone and androstenedione on the pulsatile secretion of luteinizing hormone in ovariectomized ewes during spring and autumn.Crossref | GoogleScholarGoogle Scholar | 6827203PubMed |

Martin, G. B., Durmic, Z., Kenyon, P. R., and Vercoe, P. E. (2009). Landcorp Farming Limited Lecture: “Clean, green and ethical” animal reproduction: extension to sheep and dairy systems in New Zealand. Proc. N.Z. Soc. Anim. Prod. 69, 140–147.

Martin, P., Raoul, J., and Bodin, L. (2014). Effects of the FecL major gene in the Lacaune meat sheep population. Genet. Sel. Evol. 46, 48.
Effects of the FecL major gene in the Lacaune meat sheep population.Crossref | GoogleScholarGoogle Scholar | 25158754PubMed |

Martinez, M. F., Sanderson, N., Quirke, L. D., Lawrence, S. B., and Juengel, J. L. (2016). Association between antral follicle count and reproductive measures in New Zealand lactating dairy cows maintained in a pasture-based production system. Theriogenology 85, 466–475.
Association between antral follicle count and reproductive measures in New Zealand lactating dairy cows maintained in a pasture-based production system.Crossref | GoogleScholarGoogle Scholar | 26489910PubMed |

Mathew, H., and Mahalingaiah, S. (2019). Do prenatal exposures pose a real threat to ovarian function? Bisphenol A as a case study. Reproduction 157, R143–R157.
Do prenatal exposures pose a real threat to ovarian function? Bisphenol A as a case study.Crossref | GoogleScholarGoogle Scholar | 30689546PubMed |

Mattar, D., Samir, M., Laird, M., and Knight, P. G. (2020). Modulatory effects of TGF-beta1 and BMP6 on thecal angiogenesis and steroidogenesis in the bovine ovary. Reproduction 159, 397–408.
Modulatory effects of TGF-beta1 and BMP6 on thecal angiogenesis and steroidogenesis in the bovine ovary.Crossref | GoogleScholarGoogle Scholar | 31967968PubMed |

Maylem, E. R. S., Spicer, L. J., Batalha, I., and Schutz, L. F. (2021). Discovery of a possible role of asprosin in ovarian follicular function. J. Mol. Endocrinol. 66, 35–44.
Discovery of a possible role of asprosin in ovarian follicular function.Crossref | GoogleScholarGoogle Scholar |

McClellan, K. A., Gosden, R., and Taketo, T. (2003). Continuous loss of oocytes throughout meiotic prophase in the normal mouse ovary. Dev. Biol. 258, 334–348.
Continuous loss of oocytes throughout meiotic prophase in the normal mouse ovary.Crossref | GoogleScholarGoogle Scholar | 12798292PubMed |

McNatty, K. P., Heath, D. A., Hudson, N. L., Reader, K. L., Quirke, L., Lun, S., and Juengel, J. L. (2010). The conflict between hierarchical ovarian follicular development and superovulation treatment. Reproduction 140, 287–294.
The conflict between hierarchical ovarian follicular development and superovulation treatment.Crossref | GoogleScholarGoogle Scholar | 20501789PubMed |

McNeel, A. K., and Cushman, R. A. (2015). Influence of puberty and antral follicle count on calving day in crossbred beef heifers. Theriogenology 84, 1061–1066.
Influence of puberty and antral follicle count on calving day in crossbred beef heifers.Crossref | GoogleScholarGoogle Scholar | 26197954PubMed |

McNeel, A. K., Soares, E. M., Patterson, A. L., Vallet, J. L., Wright, E. C., Larimore, E. L., Amundson, O. L., Miles, J. R., Chase, C. C., Lents, C. A., Wood, J. R., Cupp, A. S., Perry, G. A., and Cushman, R. A. (2017). Beef heifers with diminished numbers of antral follicles have decreased uterine protein concentrations. Anim. Reprod. Sci. 179, 1–9.
Beef heifers with diminished numbers of antral follicles have decreased uterine protein concentrations.Crossref | GoogleScholarGoogle Scholar | 28215453PubMed |

Mellouk, N., Rame, C., Touze, J. L., Briant, E., Ma, L., Guillaume, D., Lomet, D., Caraty, A., Ntallaris, T., Humblot, P., and Dupont, J. (2017). Involvement of plasma adipokines in metabolic and reproductive parameters in Holstein dairy cows fed with diets with differing energy levels. J. Dairy Sci. 100, 8518–8533.
Involvement of plasma adipokines in metabolic and reproductive parameters in Holstein dairy cows fed with diets with differing energy levels.Crossref | GoogleScholarGoogle Scholar | 28803009PubMed |

Monniaux, D., Baril, G., Laine, A. L., Jarrier, P., Poulin, N., Cognie, J., and Fabre, S. (2011). Anti-Mullerian hormone as a predictive endocrine marker for embryo production in the goat. Reproduction 142, 845–854.
Anti-Mullerian hormone as a predictive endocrine marker for embryo production in the goat.Crossref | GoogleScholarGoogle Scholar | 21930684PubMed |

Monniaux, D., Drouilhet, L., Rico, C., Estienne, A., Jarrier, P., Touze, J. L., Sapa, J., Phocas, F., Dupont, J., Dalbies-Tran, R., and Fabre, S. (2012). Regulation of anti-Mullerian hormone production in domestic animals. Reprod. Fertil. Dev. 25, 1–16.
Regulation of anti-Mullerian hormone production in domestic animals.Crossref | GoogleScholarGoogle Scholar | 23244824PubMed |

Monniaux, D., Clement, F., Dalbies-Tran, R., Estienne, A., Fabre, S., Mansanet, C., and Monget, P. (2014). The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link? Biol. Reprod. 90, 85.
The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link?Crossref | GoogleScholarGoogle Scholar | 24599291PubMed |

Monniaux, D., Genet, C., Maillard, V., Jarrier, P., Adriaensen, H., Hennequet-Antier, C., Laine, A. L., Laclie, C., Papillier, P., Plisson-Petit, F., Estienne, A., Cognie, J., di Clemente, N., Dalbies-Tran, R., and Fabre, S. (2020). Prenatal programming by testosterone of follicular theca cell functions in ovary. Cell. Mol. Life Sci.: CMLS 77, 1177–1196.
Prenatal programming by testosterone of follicular theca cell functions in ovary.Crossref | GoogleScholarGoogle Scholar | 31327046PubMed |

Montiel, M. D., Krzewinski-Recchi, M. A., Delannoy, P., and Harduin-Lepers, A. (2003). Molecular cloning, gene organization and expression of the human UDP-GalNAc:Neu5Acalpha2–3Galbeta-R beta1,4-N-acetylgalactosaminyltransferase responsible for the biosynthesis of the blood group Sda/Cad antigen: evidence for an unusual extended cytoplasmic domain. Biochem. J. 373, 369–379.
Molecular cloning, gene organization and expression of the human UDP-GalNAc:Neu5Acalpha2–3Galbeta-R beta1,4-N-acetylgalactosaminyltransferase responsible for the biosynthesis of the blood group Sda/Cad antigen: evidence for an unusual extended cytoplasmic domain.Crossref | GoogleScholarGoogle Scholar | 12678917PubMed |

Moraes, J. C. F., and Souza, C. J. H. (2017). Ewes carrying the Booroola and Vacaria prolificacy alleles respond differently to ovulation induction with equine chorionic gonadotrophin. Genet. Mol. Res. 16, gmr16039787.
Ewes carrying the Booroola and Vacaria prolificacy alleles respond differently to ovulation induction with equine chorionic gonadotrophin.Crossref | GoogleScholarGoogle Scholar |

Mossa, F., and Ireland, J. J. (2019). Physiology and endocrinology symposium: Anti-Mullerian hormone: a biomarker for the ovarian reserve, ovarian function, and fertility in dairy cows. J. Anim. Sci. 97, 1446–1455.
Physiology and endocrinology symposium: Anti-Mullerian hormone: a biomarker for the ovarian reserve, ovarian function, and fertility in dairy cows.Crossref | GoogleScholarGoogle Scholar | 30668706PubMed |

Mossa, F., Walsh, S. W., Butler, S. T., Berry, D. P., Carter, F., Lonergan, P., Smith, G. W., Ireland, J. J., and Evans, A. C. (2012). Low numbers of ovarian follicles >/=3 mm in diameter are associated with low fertility in dairy cows. J. Dairy Sci. 95, 2355–2361.
Low numbers of ovarian follicles >/=3 mm in diameter are associated with low fertility in dairy cows.Crossref | GoogleScholarGoogle Scholar | 22541464PubMed |

Mossa, F., Carter, F., Walsh, S. W., Kenny, D. A., Smith, G. W., Ireland, J. L., Hildebrandt, T. B., Lonergan, P., Ireland, J. J., and Evans, A. C. (2013). Maternal undernutrition in cows impairs ovarian and cardiovascular systems in their offspring. Biol. Reprod. 88, 92.
Maternal undernutrition in cows impairs ovarian and cardiovascular systems in their offspring.Crossref | GoogleScholarGoogle Scholar | 23426432PubMed |

Mossa, F., Jimenez-Krassel, F., Scheetz, D., Weber-Nielsen, M., Evans, A. C. O., and Ireland, J. J. (2017). Anti-Mullerian Hormone (AMH) and fertility management in agricultural species. Reproduction 154, R1–R11.
Anti-Mullerian Hormone (AMH) and fertility management in agricultural species.Crossref | GoogleScholarGoogle Scholar | 28356501PubMed |

Murdoch, W. J., Van Kirk, E. A., Vonnahme, K. A., and Ford, S. P. (2003). Ovarian responses to undernutrition in pregnant ewes, USA. Reprod. Biol. Endocrinol. 1, 6.
Ovarian responses to undernutrition in pregnant ewes, USA.Crossref | GoogleScholarGoogle Scholar | 12646075PubMed |

Murray, L. A. (2019). Refinement of a physiologically-relevant IVM/IVF system on oocyte developmental competency in New Zealand dairy cows. PhD thesis, Victoria University of Wellington, New Zealand.

Nawaz, M. Y., Jimenez-Krassel, F., Steibel, J. P., Lu, Y., Baktula, A., Vukasinovic, N., Neuder, L., Ireland, J. L. H., Ireland, J. J., and Tempelman, R. J. (2018). Genomic heritability and genome-wide association analysis of anti-Mullerian hormone in Holstein dairy heifers. J. Dairy Sci. 101, 8063–8075.
Genomic heritability and genome-wide association analysis of anti-Mullerian hormone in Holstein dairy heifers.Crossref | GoogleScholarGoogle Scholar | 30007805PubMed |

Nichols, J. A., Perego, M. C., Schutz, L. F., Hemple, A. M., and Spicer, L. J. (2019). Hormonal regulation of vascular endothelial growth factor A (VEGFA) gene expression in granulosa and theca cells of cattle1. J. Anim. Sci. 97, 3034–3045.
Hormonal regulation of vascular endothelial growth factor A (VEGFA) gene expression in granulosa and theca cells of cattle1.Crossref | GoogleScholarGoogle Scholar | 31077271PubMed |

Norris, R. P., Freudzon, M., Mehlmann, L. M., Cowan, A. E., Simon, A. M., Paul, D. L., Lampe, P. D., and Jaffe, L. A. (2008). Luteinizing hormone causes MAP kinase-dependent phosphorylation and closure of connexin 43 gap junctions in mouse ovarian follicles: one of two paths to meiotic resumption. Development 135, 3229–3238.
Luteinizing hormone causes MAP kinase-dependent phosphorylation and closure of connexin 43 gap junctions in mouse ovarian follicles: one of two paths to meiotic resumption.Crossref | GoogleScholarGoogle Scholar | 18776144PubMed |

O’Shea, T., Hillard, M. A., Anderson, S. T., Bindon, B. M., Findlay, J. K., Tsonis, C. G., and Wilkins, J. F. (1994). Inhibin immunization for increasing ovulation rate and superovulation. Theriogenology 41, 3–17.
Inhibin immunization for increasing ovulation rate and superovulation.Crossref | GoogleScholarGoogle Scholar |

Oliveira Júnior, G. A., Perez, B. C., Cole, J. B., Santana, M. H. A., Silveira, J., Mazzoni, G., Ventura, R. V., Santana, M. L. J., Kadarmideen, H. N., Garrick, D. J., and Ferraz, J. B. S. (2017). Genomic study and Medical Subject Headings enrichment analysis of early pregnancy rate and antral follicle numbers in Nelore heifers. J. Anim. Sci. 95, 4796–4812.
Genomic study and Medical Subject Headings enrichment analysis of early pregnancy rate and antral follicle numbers in Nelore heifers.Crossref | GoogleScholarGoogle Scholar |

Padmanabhan, V., and Veiga-Lopez, A. (2013). Sheep models of polycystic ovary syndrome phenotype. Mol. Cell. Endocrinol. 373, 8–20.
Sheep models of polycystic ovary syndrome phenotype.Crossref | GoogleScholarGoogle Scholar | 23084976PubMed |

Pan, Z., Zhang, J., Li, Q., Li, Y., Shi, F., Xie, Z., and Liu, H. (2012). Current advances in epigenetic modification and alteration during mammalian ovarian folliculogenesis. J. Genet. Genomics 39, 111–123.
Current advances in epigenetic modification and alteration during mammalian ovarian folliculogenesis.Crossref | GoogleScholarGoogle Scholar | 22464470PubMed |

Parrott, J. A., and Skinner, M. K. (1998). Developmental and hormonal regulation of keratinocyte growth factor expression and action in the ovarian follicle. Endocrinology 139, 228–235.
Developmental and hormonal regulation of keratinocyte growth factor expression and action in the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 9421419PubMed |

Pfeiffer, K. E., Jury, L. J., and Larson, J. E. (2014). Determination of anti-Mullerian hormone at estrus during a synchronized and a natural bovine estrous cycle. Domest. Anim. Endocrinol. 46, 58–64.
Determination of anti-Mullerian hormone at estrus during a synchronized and a natural bovine estrous cycle.Crossref | GoogleScholarGoogle Scholar | 24211073PubMed |

Pierre, A., Estienne, A., Racine, C., Picard, J. Y., Fanchin, R., Lahoz, B., Alabart, J. L., Folch, J., Jarrier, P., Fabre, S., Monniaux, D., and di Clemente, N. (2016). The bone morphogenetic protein 15 up-regulates the anti-Mullerian hormone receptor expression in granulosa cells. J. Clin. Endocrinol. Metab. 101, 2602–2611.
The bone morphogenetic protein 15 up-regulates the anti-Mullerian hormone receptor expression in granulosa cells.Crossref | GoogleScholarGoogle Scholar | 27070094PubMed |

Pinto, P. H. N., Balaro, M. F. A., Souza-Fabjan, J. M. G., Ribeiro, L. D. S., Braganca, G. M., Leite, C. R., Arashiro, E. K. N., de Moraes Silva, K., Da Fonseca, J. F., and Brandao, F. Z. (2018). Anti-Mullerian hormone and antral follicle count are more effective for selecting ewes with good potential for in vivo embryo production than the presence of FecG(E) mutation or eCG pre-selection tests. Theriogenology 113, 146–152.
Anti-Mullerian hormone and antral follicle count are more effective for selecting ewes with good potential for in vivo embryo production than the presence of FecG(E) mutation or eCG pre-selection tests.Crossref | GoogleScholarGoogle Scholar |

Pocar, P., Brevini, T. A., Perazzoli, F., Cillo, F., Modina, S., and Gandolfi, F. (2001). Cellular and molecular mechanisms mediating the effects of polychlorinated biphenyls on oocyte developmental competence in cattle. Mol. Reprod. Dev. 60, 535–541.
Cellular and molecular mechanisms mediating the effects of polychlorinated biphenyls on oocyte developmental competence in cattle.Crossref | GoogleScholarGoogle Scholar | 11746964PubMed |

Poole, D. H., Ocon-Grove, O. M., and Johnson, A. L. (2016). Anti-Mullerian hormone (AMH) receptor type II expression and AMH activity in bovine granulosa cells. Theriogenology 86, 1353–1360.
Anti-Mullerian hormone (AMH) receptor type II expression and AMH activity in bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 27268296PubMed |

Portela, V. M., Dirandeh, E., Guerrero-Netro, H. M., Zamberlam, G., Barreta, M. H., Goetten, A. F., and Price, C. A. (2015). The role of fibroblast growth factor-18 in follicular atresia in cattle. Biol. Reprod. 92, 14.
The role of fibroblast growth factor-18 in follicular atresia in cattle.Crossref | GoogleScholarGoogle Scholar | 25411391PubMed |

Price, C. A. (2016). Mechanisms of fibroblast growth factor signaling in the ovarian follicle. J. Endocrinol. 228, R31–R43.
Mechanisms of fibroblast growth factor signaling in the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 26542145PubMed |

Qi, M. Y., Xu, L. Q., Zhang, J. N., Li, M. O., Lu, M. H., and Yao, Y. C. (2020). Effect of the Booroola fecundity (FecB) gene on the reproductive performance of ewes under assisted reproduction. Theriogenology 142, 246–250.
Effect of the Booroola fecundity (FecB) gene on the reproductive performance of ewes under assisted reproduction.Crossref | GoogleScholarGoogle Scholar | 31711699PubMed |

Rae, M. T., Palassio, S., Kyle, C. E., Brooks, A. N., Lea, R. G., Miller, D. W., and Rhind, S. M. (2001). Effect of maternal undernutrition during pregnancy on early ovarian development and subsequent follicular development in sheep fetuses. Reproduction 122, 915–922.
Effect of maternal undernutrition during pregnancy on early ovarian development and subsequent follicular development in sheep fetuses.Crossref | GoogleScholarGoogle Scholar | 11732987PubMed |

Rawlings, N. C., Evans, A. C., Honaramooz, A., and Bartlewski, P. M. (2003). Antral follicle growth and endocrine changes in prepubertal cattle, sheep and goats. Anim. Reprod. Sci. 78, 259–270.
Antral follicle growth and endocrine changes in prepubertal cattle, sheep and goats.Crossref | GoogleScholarGoogle Scholar | 12818648PubMed |

Reverchon, M., Rame, C., Cognie, J., Briant, E., Elis, S., Guillaume, D., and Dupont, J. (2014). Resistin in dairy cows: plasma concentrations during early lactation, expression and potential role in adipose tissue. PLoS One 9, e93198.
Resistin in dairy cows: plasma concentrations during early lactation, expression and potential role in adipose tissue.Crossref | GoogleScholarGoogle Scholar | 24675707PubMed |

Reverchon, M., Rame, C., Bunel, A., Chen, W., Froment, P., and Dupont, J. (2016). VISFATIN (NAMPT) improves in vitro IGF1-induced steroidogenesis and IGF1 receptor signaling through SIRT1 in bovine granulosa cells. Biol. Reprod. 94, 54.
VISFATIN (NAMPT) improves in vitro IGF1-induced steroidogenesis and IGF1 receptor signaling through SIRT1 in bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 26792944PubMed |

Rhind, S. M., Smith, A., Kyle, C. E., Telfer, G., Martin, G., Duff, E., and Mayes, R. W. (2002). Phthalate and alkyl phenol concentrations in soil following applications of inorganic fertiliser or sewage sludge to pasture and potential rates of ingestion by grazing ruminants. J. Environ. Monit. 4, 142–148.
Phthalate and alkyl phenol concentrations in soil following applications of inorganic fertiliser or sewage sludge to pasture and potential rates of ingestion by grazing ruminants.Crossref | GoogleScholarGoogle Scholar | 11871695PubMed |

Rhind, S. M., Kyle, C. E., Telfer, G., Duff, E. I., and Smith, A. (2005). Alkyl phenols and diethylhexyl phthalate in tissues of sheep grazing pastures fertilized with sewage sludge or inorganic fertilizer. Environ. Health Perspect. 113, 447–453.
Alkyl phenols and diethylhexyl phthalate in tissues of sheep grazing pastures fertilized with sewage sludge or inorganic fertilizer.Crossref | GoogleScholarGoogle Scholar | 15811823PubMed |

Rhind, S. M., Kyle, C. E., Mackie, C., and McDonald, L. (2009). Accumulation of endocrine disrupting compounds in sheep fetal and maternal liver tissue following exposure to pastures treated with sewage sludge. J. Environ. Monit. 11, 1469–1476.
Accumulation of endocrine disrupting compounds in sheep fetal and maternal liver tissue following exposure to pastures treated with sewage sludge.Crossref | GoogleScholarGoogle Scholar | 19657530PubMed |

Rhind, S. M., Kyle, C. E., Mackie, C., McDonald, L., Zhang, Z., Duff, E. I., Bellingham, M., Amezaga, M. R., Mandon-Pepin, B., Loup, B., Cotinot, C., Evans, N. P., Sharpe, R. M., and Fowler, P. A. (2010). Maternal and fetal tissue accumulation of selected endocrine disrupting compounds (EDCs) following exposure to sewage sludge-treated pastures before or after conception. J. Environ. Monit. 12, 1582–1593.
Maternal and fetal tissue accumulation of selected endocrine disrupting compounds (EDCs) following exposure to sewage sludge-treated pastures before or after conception.Crossref | GoogleScholarGoogle Scholar | 20676422PubMed |

Rhind, S. M., Kyle, C. E., Ruffie, H., Calmettes, E., Osprey, M., Zhang, Z. L., Hamilton, D., and McKenzie, C. (2013). Short- and long-term temporal changes in soil concentrations of selected endocrine disrupting compounds (EDCs) following single or multiple applications of sewage sludge to pastures. Environ. Pollut. 181, 262–270.
Short- and long-term temporal changes in soil concentrations of selected endocrine disrupting compounds (EDCs) following single or multiple applications of sewage sludge to pastures.Crossref | GoogleScholarGoogle Scholar | 23896644PubMed |

Rico, C., Fabre, S., Medigue, C., di Clemente, N., Clement, F., Bontoux, M., Touze, J. L., Dupont, M., Briant, E., Remy, B., Beckers, J. F., and Monniaux, D. (2009). Anti-mullerian hormone is an endocrine marker of ovarian gonadotropin-responsive follicles and can help to predict superovulatory responses in the cow. Biol. Reprod. 80, 50–59.
Anti-mullerian hormone is an endocrine marker of ovarian gonadotropin-responsive follicles and can help to predict superovulatory responses in the cow.Crossref | GoogleScholarGoogle Scholar | 18784351PubMed |

Rico, C., Medigue, C., Fabre, S., Jarrier, P., Bontoux, M., Clement, F., and Monniaux, D. (2011). Regulation of anti-Mullerian hormone production in the cow: a multiscale study at endocrine, ovarian, follicular, and granulosa cell levels. Biol. Reprod. 84, 560–571.
Regulation of anti-Mullerian hormone production in the cow: a multiscale study at endocrine, ovarian, follicular, and granulosa cell levels.Crossref | GoogleScholarGoogle Scholar | 21076084PubMed |

Rivera, O. E., Varayoud, J., Rodriguez, H. A., Munoz-de-Toro, M., and Luque, E. H. (2011). Neonatal exposure to bisphenol A or diethylstilbestrol alters the ovarian follicular dynamics in the lamb. Reprod. Toxicol. 32, 304–312.
Neonatal exposure to bisphenol A or diethylstilbestrol alters the ovarian follicular dynamics in the lamb.Crossref | GoogleScholarGoogle Scholar | 21722727PubMed |

Roche, J., Rame, C., Reverchon, M., Mellouk, N., Rak, A., Froment, P., and Dupont, J. (2017). Apelin (APLN) regulates progesterone secretion and oocyte maturation in bovine ovarian cells. Reproduction 153, 589–603.
Apelin (APLN) regulates progesterone secretion and oocyte maturation in bovine ovarian cells.Crossref | GoogleScholarGoogle Scholar | 28250234PubMed |

Samir, M., Glister, C., Mattar, D., Laird, M., and Knight, P. G. (2017). Follicular expression of pro-inflammatory cytokines tumour necrosis factor-alpha (TNFalpha), interleukin 6 (IL6) and their receptors in cattle: TNFalpha, IL6 and macrophages suppress thecal androgen production in vitro. Reproduction 154, 35–49.
Follicular expression of pro-inflammatory cytokines tumour necrosis factor-alpha (TNFalpha), interleukin 6 (IL6) and their receptors in cattle: TNFalpha, IL6 and macrophages suppress thecal androgen production in vitro.Crossref | GoogleScholarGoogle Scholar | 28432091PubMed |

Santa Cruz, R., Cushman, R. A., and Vinoles, C. (2018). Antral follicular count is a tool that may allow the selection of more precocious Bradford heifers at weaning. Theriogenology 119, 35–42.
Antral follicular count is a tool that may allow the selection of more precocious Bradford heifers at weaning.Crossref | GoogleScholarGoogle Scholar | 29982134PubMed |

Santiquet, N. W., Greene, A. F., Becker, J., Barfield, J. P., Schoolcraft, W. B., and Krisher, R. L. (2017). A pre-in vitro maturation medium containing cumulus oocyte complex ligand-receptor signaling molecules maintains meiotic arrest, supports the cumulus oocyte complex and improves oocyte developmental competence. Mol. Hum. Reprod. 23, 594–606.
A pre-in vitro maturation medium containing cumulus oocyte complex ligand-receptor signaling molecules maintains meiotic arrest, supports the cumulus oocyte complex and improves oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 28586460PubMed |

Savabieasfahani, M., Kannan, K., Astapova, O., Evans, N. P., and Padmanabhan, V. (2006). Developmental programming: differential effects of prenatal exposure to bisphenol-A or methoxychlor on reproductive function. Endocrinology 147, 5956–5966.
Developmental programming: differential effects of prenatal exposure to bisphenol-A or methoxychlor on reproductive function.Crossref | GoogleScholarGoogle Scholar | 16946013PubMed |

Sawyer, H. R., Smith, P., Heath, D. A., Juengel, J. L., Wakefield, S. J., and McNatty, K. P. (2002). Formation of ovarian follicles during fetal development in sheep. Biol. Reprod. 66, 1134–1150.
Formation of ovarian follicles during fetal development in sheep.Crossref | GoogleScholarGoogle Scholar | 11906935PubMed |

Scaramuzzi, R. J., Adams, N. R., Baird, D. T., Campbell, B. K., Downing, J. A., Findlay, J. K., Henderson, K. M., Martin, G. B., McNatty, K. P., McNeilly, A. S., and Tsonis, C. G. (1993). A model for follicle selection and the determination of ovulation rate in the ewe. Reprod. Fertil. Dev. 5, 459–478.
A model for follicle selection and the determination of ovulation rate in the ewe.Crossref | GoogleScholarGoogle Scholar | 8190903PubMed |

Scaramuzzi, R. J., Baird, D. T., Campbell, B. K., Driancourt, M. A., Dupont, J., Fortune, J. E., Gilchrist, R. B., Martin, G. B., McNatty, K. P., McNeilly, A. S., Monget, P., Monniaux, D., Vinoles, C., and Webb, R. (2011). Regulation of folliculogenesis and the determination of ovulation rate in ruminants. Reprod. Fertil. Dev. 23, 444–467.
Regulation of folliculogenesis and the determination of ovulation rate in ruminants.Crossref | GoogleScholarGoogle Scholar | 21426863PubMed |

Scheetz, D., Folger, J. K., Smith, G. W., and Ireland, J. J. (2012). Granulosa cells are refractory to FSH action in individuals with a low antral follicle count. Reprod. Fertil. Dev. 24, 327–336.
Granulosa cells are refractory to FSH action in individuals with a low antral follicle count.Crossref | GoogleScholarGoogle Scholar | 22281079PubMed |

Sela-Abramovich, S., Edry, I., Galiani, D., Nevo, N., and Dekel, N. (2006). Disruption of gap junctional communication within the ovarian follicle induces oocyte maturation. Endocrinology 147, 2280–2286.
Disruption of gap junctional communication within the ovarian follicle induces oocyte maturation.Crossref | GoogleScholarGoogle Scholar | 16439460PubMed |

Sharma, B., Sarkar, A., Singh, P., and Singh, R. P. (2017). Agricultural utilization of biosolids: A review on potential effects on soil and plant grown. Waste Manag. 64, 117–132.
Agricultural utilization of biosolids: A review on potential effects on soil and plant grown.Crossref | GoogleScholarGoogle Scholar | 28336334PubMed |

Shimizu, T. (2016). Molecular and cellular mechanisms for the regulation of ovarian follicular function in cows. J. Reprod. Dev. 62, 323–329.
Molecular and cellular mechanisms for the regulation of ovarian follicular function in cows.Crossref | GoogleScholarGoogle Scholar | 27097851PubMed |

Siemienowicz, K., Rae, M. T., Howells, F., Anderson, C., Nicol, L. M., Franks, S., and Duncan, W. C. (2020). Insights into manipulating postprandial energy expenditure to manage weight gain in polycystic ovary syndrome. iScience 23, 101164.
Insights into manipulating postprandial energy expenditure to manage weight gain in polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 32464593PubMed |

Sinclair, K. D., Rutherford, K. M., Wallace, J. M., Brameld, J. M., Stoger, R., Alberio, R., Sweetman, D., Gardner, D. S., Perry, V. E., Adam, C. L., Ashworth, C. J., Robinson, J. E., and Dwyer, C. M. (2016). Epigenetics and developmental programming of welfare and production traits in farm animals. Reprod. Fertil. Dev. 28, 1443–1478.
Epigenetics and developmental programming of welfare and production traits in farm animals.Crossref | GoogleScholarGoogle Scholar |

Smith, P., Steckler, T. L., Veiga-Lopez, A., and Padmanabhan, V. (2009). Developmental programming: differential effects of prenatal testosterone and dihydrotestosterone on follicular recruitment, depletion of follicular reserve, and ovarian morphology in sheep. Biol. Reprod. 80, 726–736.
Developmental programming: differential effects of prenatal testosterone and dihydrotestosterone on follicular recruitment, depletion of follicular reserve, and ovarian morphology in sheep.Crossref | GoogleScholarGoogle Scholar | 19092114PubMed |

Smith, P., Wilhelm, D., and Rodgers, R. J. (2014). Development of mammalian ovary. J. Endocrinol. 221, R145–R161.
Development of mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 24741072PubMed |

Smith, P., Juengel, J., Maclean, P., Rand, C., and Stanton, J. L. (2019a). Gestational nutrition 2: gene expression in sheep fetal ovaries exposed to gestational under nutrition. Reproduction 157, 13–25.
Gestational nutrition 2: gene expression in sheep fetal ovaries exposed to gestational under nutrition.Crossref | GoogleScholarGoogle Scholar | 30394704PubMed |

Smith, P., Stanton, J. L., Quirke, L., and Juengel, J. L. (2019b). Gestational nutrition 1: alterations to gestational nutrition can increase indicators of fertility in sheep. Reproduction 157, 199–213.
Gestational nutrition 1: alterations to gestational nutrition can increase indicators of fertility in sheep.Crossref | GoogleScholarGoogle Scholar | 30817311PubMed |

Snelling, W. M., Cushman, R. A., Fortes, M. R., Reverter, A., Bennett, G. L., Keele, J. W., Kuehn, L. A., McDaneld, T. G., Thallman, R. M., and Thomas, M. G. (2012). Physiology and Endocrinology Symposium: How single nucleotide polymorphism chips will advance our knowledge of factors controlling puberty and aid in selecting replacement beef females. J. Anim. Sci. 90, 1152–1165.
Physiology and Endocrinology Symposium: How single nucleotide polymorphism chips will advance our knowledge of factors controlling puberty and aid in selecting replacement beef females.Crossref | GoogleScholarGoogle Scholar | 22038989PubMed |

Spicer, L. J., Aad, P. Y., Allen, D. T., Mazerbourg, S., Payne, A. H., and Hsueh, A. J. (2008). Growth differentiation factor 9 (GDF9) stimulates proliferation and inhibits steroidogenesis by bovine theca cells: influence of follicle size on responses to GDF9. Biol. Reprod. 78, 243–253.
Growth differentiation factor 9 (GDF9) stimulates proliferation and inhibits steroidogenesis by bovine theca cells: influence of follicle size on responses to GDF9.Crossref | GoogleScholarGoogle Scholar | 17959852PubMed |

Spicer, L. J., Schreiber, N. B., Lagaly, D. V., Aad, P. Y., Douthit, L. B., and Grado-Ahuir, J. A. (2011). Effect of resistin on granulosa and theca cell function in cattle. Anim. Reprod. Sci. 124, 19–27.
Effect of resistin on granulosa and theca cell function in cattle.Crossref | GoogleScholarGoogle Scholar | 21315524PubMed |

Su, Y. Q., Sugiura, K., Wigglesworth, K., O’Brien, M. J., Affourtit, J. P., Pangas, S. A., Matzuk, M. M., and Eppig, J. J. (2008). Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. Development 135, 111–121.
Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells.Crossref | GoogleScholarGoogle Scholar | 18045843PubMed |

Succu, S., Sale, S., Ghirello, G., Ireland, J. J., Evans, A. C. O., Atzori, A. S., and Mossa, F. (2020). Exposure of dairy cows to high environmental temperatures and their lactation status impairs establishment of the ovarian reserve in their offspring. J. Dairy Sci. 103, 11957–11969.
Exposure of dairy cows to high environmental temperatures and their lactation status impairs establishment of the ovarian reserve in their offspring.Crossref | GoogleScholarGoogle Scholar | 33041040PubMed |

Sugimura, S., Ritter, L. J., Rose, R. D., Thompson, J. G., Smitz, J., Mottershead, D. G., and Gilchrist, R. B. (2015). Promotion of EGF receptor signaling improves the quality of low developmental competence oocytes. Dev. Biol. 403, 139–149.
Promotion of EGF receptor signaling improves the quality of low developmental competence oocytes.Crossref | GoogleScholarGoogle Scholar | 25981108PubMed |

Susiarjo, M., Hassold, T. J., Freeman, E., and Hunt, P. A. (2007). Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genet. 3, e5.
Bisphenol A exposure in utero disrupts early oogenesis in the mouse.Crossref | GoogleScholarGoogle Scholar | 17222059PubMed |

Tabandeh, M. R., Hosseini, A., Saeb, M., Kafi, M., and Saeb, S. (2010). Changes in the gene expression of adiponectin and adiponectin receptors (AdipoR1 and AdipoR2) in ovarian follicular cells of dairy cow at different stages of development. Theriogenology 73, 659–669.
Changes in the gene expression of adiponectin and adiponectin receptors (AdipoR1 and AdipoR2) in ovarian follicular cells of dairy cow at different stages of development.Crossref | GoogleScholarGoogle Scholar | 20047754PubMed |

Tay, S. H., Blache, D., Gregg, K., and Revell, D. K. (2012). Consumption of a high-salt diet by ewes during pregnancy alters nephrogenesis in 5-month-old offspring. Animal 6, 1803–1810.
Consumption of a high-salt diet by ewes during pregnancy alters nephrogenesis in 5-month-old offspring.Crossref | GoogleScholarGoogle Scholar |

Tenley, S. C., Gomes, R. S., Rosasco, S. L., Northrop, E. J., Rich, J. J. J., McNeel, A. K., Summers, A. F., Miles, J. R., Chase, C. C., Lents, C. A., Perry, G. A., Wood, J. R., Cupp, A. S., and Cushman, R. A. (2019). Maternal age influences the number of primordial follicles in the ovaries of yearling Angus heifers. Anim. Reprod. Sci. 200, 105–112.
Maternal age influences the number of primordial follicles in the ovaries of yearling Angus heifers.Crossref | GoogleScholarGoogle Scholar | 30563721PubMed |

Torres-Rovira, L., Gonzalez-Bulnes, A., Succu, S., Spezzigu, A., Manca, M. E., Leoni, G. G., Sanna, M., Pirino, S., Gallus, M., Naitana, S., and Berlinguer, F. (2014). Predictive value of antral follicle count and anti-Mullerian hormone for follicle and oocyte developmental competence during the early prepubertal period in a sheep model. Reprod. Fertil. Dev. 26, 1094–1106.
Predictive value of antral follicle count and anti-Mullerian hormone for follicle and oocyte developmental competence during the early prepubertal period in a sheep model.Crossref | GoogleScholarGoogle Scholar | 24008140PubMed |

Tsafriri, A., and Dekel, N. (1994) Molecular mechanisms in ovulation. In ‘Molecular Biology of the Female Reproductive System’. (Ed. J. K. Findlay) pp. 207–258. (Academic Press: San Diego)

Turner, E. C., Hughes, J., Wilson, H., Clay, M., Mylonas, K. J., Kipari, T., Duncan, W. C., and Fraser, H. M. (2011). Conditional ablation of macrophages disrupts ovarian vasculature. Reproduction 141, 821–831.
Conditional ablation of macrophages disrupts ovarian vasculature.Crossref | GoogleScholarGoogle Scholar | 21393340PubMed |

Umer, S., Sammad, A., Zou, H., Khan, A., Weldegebriall Sahlu, B., Hao, H., Zhao, X., Wang, Y., Zhao, S., and Zhu, H. (2019). Regulation of AMH, AMHR-II, and BMPs (2,6) genes of bovine granulosa cells treated with exogenous fSH and their association with protein hormones. Genes (Basel) 10, 1038.
Regulation of AMH, AMHR-II, and BMPs (2,6) genes of bovine granulosa cells treated with exogenous fSH and their association with protein hormones.Crossref | GoogleScholarGoogle Scholar |

Vaccari, S., Weeks, J. L., Hsieh, M., Menniti, F. S., and Conti, M. (2009). Cyclic GMP signaling is involved in the luteinizing hormone-dependent meiotic maturation of mouse oocytes. Biol. Reprod. 81, 595–604.
Cyclic GMP signaling is involved in the luteinizing hormone-dependent meiotic maturation of mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 19474061PubMed |

Van Vleck, L. D., Gregory, K. E., and Echternkamp, S. E. (1991). Ovulation rate and twinning rate in cattle: heritabilities and genetic correlation. J. Anim. Sci. 69, 3213–3219.
Ovulation rate and twinning rate in cattle: heritabilities and genetic correlation.Crossref | GoogleScholarGoogle Scholar | 1894556PubMed |

Veiga-Lopez, A., Luense, L. J., Christenson, L. K., and Padmanabhan, V. (2013). Developmental programming: gestational bisphenol-A treatment alters trajectory of fetal ovarian gene expression. Endocrinology 154, 1873–1884.
Developmental programming: gestational bisphenol-A treatment alters trajectory of fetal ovarian gene expression.Crossref | GoogleScholarGoogle Scholar | 23525218PubMed |

Vigier, B., Picard, J. Y., Tran, D., Legeai, L., and Josso, N. (1984). Production of anti-Mullerian hormone: another homology between Sertoli and granulosa cells. Endocrinology 114, 1315–1320.
Production of anti-Mullerian hormone: another homology between Sertoli and granulosa cells.Crossref | GoogleScholarGoogle Scholar | 6546716PubMed |

Walsh, S. W., Mossa, F., Butler, S. T., Berry, D. P., Scheetz, D., Jimenez-Krassel, F., Tempelman, R. J., Carter, F., Lonergan, P., Evans, A. C., and Ireland, J. J. (2014). Heritability and impact of environmental effects during pregnancy on antral follicle count in cattle. J. Dairy Sci. 97, 4503–4511.
Heritability and impact of environmental effects during pregnancy on antral follicle count in cattle.Crossref | GoogleScholarGoogle Scholar | 24835969PubMed |

Wartenberg, H., Ihmer, A., Schwarz, S., Miething, A., and Viebahn, C. (2001). Mitotic arrest of female germ cells during prenatal oogenesis. A colcemid-like, non-apoptotic cell death. Anat. Embryol. (Berl.) 204, 421–435.
Mitotic arrest of female germ cells during prenatal oogenesis. A colcemid-like, non-apoptotic cell death.Crossref | GoogleScholarGoogle Scholar | 11789990PubMed |

Webb, R., and Campbell, B. K. (2007). Development of the dominant follicle: Mechanisms of selection and maintenance of oocyte quality. Soc. Reprod. Fertil. Suppl. 64, 141–163.
Development of the dominant follicle: Mechanisms of selection and maintenance of oocyte quality.Crossref | GoogleScholarGoogle Scholar | 17491145PubMed |

Wettemann, R. P., Lents, C. A., Ciccioli, N. H., White, F. J., and Rubio, I. (2003). Nutritional- and suckling-mediated anovulation in beef cows. J. Anim. Sci. 81, E48–E59.
Nutritional- and suckling-mediated anovulation in beef cows.Crossref | GoogleScholarGoogle Scholar |

Wigglesworth, K., Lee, K. B., O’Brien, M. J., Peng, J., Matzuk, M. M., and Eppig, J. J. (2013). Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. Proc. Natl. Acad. Sci. USA 110, E3723–E3729.
Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes.Crossref | GoogleScholarGoogle Scholar | 23980176PubMed |

Yang, M. Y., Cushman, R. A., and Fortune, J. E. (2017). Anti-Mullerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles. Mol. Hum. Reprod. 23, 282–291.
Anti-Mullerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles.Crossref | GoogleScholarGoogle Scholar | 28333275PubMed |

Young, J. M., and McNeilly, A. S. (2010). Theca: the forgotten cell of the ovarian follicle. Reproduction 140, 489–504.
Theca: the forgotten cell of the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 20628033PubMed |

Zeng, Y., and Chen, T. (2019). DNA methylation reprogramming during mammalian development. Genes (Basel) 10, 257.
DNA methylation reprogramming during mammalian development.Crossref | GoogleScholarGoogle Scholar |

Zhang, M., Su, Y. Q., Sugiura, K., Xia, G., and Eppig, J. J. (2010). Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science 330, 366–369.
Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 20947764PubMed |

Zhang, M., Su, Y. Q., Sugiura, K., Wigglesworth, K., Xia, G., and Eppig, J. J. (2011). Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro. Endocrinology 152, 4377–4385.
Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro.Crossref | GoogleScholarGoogle Scholar | 21914782PubMed |

Zhang, M., Jiang, M., Bi, Y., Zhu, H., Zhou, Z., and Sha, J. (2012). Autophagy and apoptosis act as partners to induce germ cell death after heat stress in mice. PLoS One 7, e41412.
Autophagy and apoptosis act as partners to induce germ cell death after heat stress in mice.Crossref | GoogleScholarGoogle Scholar | 23285222PubMed |

Zhang, T., Zhang, C., Fan, X., Li, R., and Zhang, J. (2017a). Effect of C-type natriuretic peptide pretreatment on in vitro bovine oocyte maturation. In vitro Cell. Dev. Biol. Anim. 53, 199–206.
Effect of C-type natriuretic peptide pretreatment on in vitro bovine oocyte maturation.Crossref | GoogleScholarGoogle Scholar | 27761784PubMed |

Zhang, Y., Li, F., Feng, X., Yang, H., Zhu, A., Pang, J., Han, L., Zhang, T., Yao, X., and Wang, F. (2017b). Genome-wide analysis of DNA methylation profiles on sheep ovaries associated with prolificacy using whole-genome bisulfite sequencing. BMC Genomics 18, 759.
Genome-wide analysis of DNA methylation profiles on sheep ovaries associated with prolificacy using whole-genome bisulfite sequencing.Crossref | GoogleScholarGoogle Scholar | 28969601PubMed |

Zhao, Z., Guo, F., Sun, X., He, Q., Dai, Z., Chen, X., Zhao, Y., and Wang, J. (2018). BMP15 regulates AMH expression via the p38 MAPK pathway in granulosa cells from goat. Theriogenology 118, 72–79.
BMP15 regulates AMH expression via the p38 MAPK pathway in granulosa cells from goat.Crossref | GoogleScholarGoogle Scholar | 29885643PubMed |