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

Effect of superstimulatory treatments on the expression of genes related to ovulatory capacity, oocyte competence and embryo development in cattle

Ciro M. Barros A C , Rafael A. Satrapa A , Anthony C. S. Castilho A , Patrícia K. Fontes A , Eduardo M. Razza A , Ronaldo L. Ereno A and Marcelo F. G. Nogueira B
+ Author Affiliations
- Author Affiliations

A Department of Pharmacology, Institute of Bioscience, University of Sao Paulo State (UNESP), Rubiao Jr S/N, Botucatu, 18618-970, SP, Brazil.

B Department of Biological Science, University of Sao Paulo State (UNESP), Assis, 19806-900, SP, Brazil.

C Corresponding author. Email: cmbarros@ibb.unesp.br

Reproduction, Fertility and Development 25(1) 17-25 https://doi.org/10.1071/RD12271
Published: 4 December 2012

Abstract

Multiple ovulation (superovulation) and embryo transfer has been used extensively in cattle. In the past decade, superstimulatory treatment protocols that synchronise follicle growth and ovulation, allowing for improved donor management and fixed-time AI (FTAI), have been developed for zebu (Bos indicus) and European (Bos taurus) breeds of cattle. There is evidence that additional stimulus with LH (through the administration of exogenous LH or equine chorionic gonadotrophin (eCG)) on the last day of the superstimulatory treatment protocol, called the ‘P-36 protocol’ for FTAI, can increase embryo yield compared with conventional protocols that are based on the detection of oestrus. However, inconsistent results with the use of hormones that stimulate LH receptors (LHR) have prompted further studies on the roles of LH and its receptors in ovulatory capacity (acquisition of LHR in granulosa cells), oocyte competence and embryo quality in superstimulated cattle. Recent experiments have shown that superstimulation with FSH increases mRNA expression of LHR and angiotensin AT2 receptors in granulosa cells of follicles >8 mm in diameter. In addition, FSH decreases mRNA expression of growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) in oocytes, but increases the expression of both in cumulus cells, without diminishing the capacity of cumulus–oocyte complexes to generate blastocysts. Although these results indicate that superstimulation with FSH is not detrimental to oocyte competence, supplementary studies are warranted to investigate the effects of superstimulation on embryo quality and viability. In addition, experiments comparing the cellular and/or molecular effects of adding eCG to the P-36 treatment protocol are being conducted to elucidate the effects of superstimulatory protocols on the yield of viable embryos.

Additional keywords: bovine, embryo transfer, FSH, LH, LH receptor.


References

Adriaens, I., Cortvrindt, R., and Smitz, J. (2004). Differential FSH exposure in preantral follicle culture has marked effects on folliculogenesis and oocyte developmental competence. Hum. Reprod. 19, 398–408.
Differential FSH exposure in preantral follicle culture has marked effects on folliculogenesis and oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXos1entQ%3D%3D&md5=2e95f9ab35b3abd0a8a4aaadcfaba41dCAS |

Ali, A., and Sirard, M. A. (2005). Protein kinases influence bovine oocyte competence during short-term treatment with recombinant human follicle stimulating hormone. Reproduction 130, 303–310.
Protein kinases influence bovine oocyte competence during short-term treatment with recombinant human follicle stimulating hormone.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtFWisr3M&md5=2119e228e7d2808d9667dd1d875dc4dcCAS |

Ascoli, M., Fanelli, F., and Segaloff, D. L. (2002). The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr. Rev. 23, 141–174.
The lutropin/choriogonadotropin receptor, a 2002 perspective.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XjsFWjtr4%3D&md5=08e4e98e9df1294bd08184653aa7d2b6CAS |

Atger, M., Misrahi, M., Sar, S., Le Flem, L., Dessen, P., and Milgrom, E. (1995). Structure of the human luteinizing hormone–choriogonadotropin receptor gene: unusual promoter and 5′ non-coding regions. Mol. Cell. Endocrinol. 111, 113–123.
Structure of the human luteinizing hormone–choriogonadotropin receptor gene: unusual promoter and 5′ non-coding regions.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXmsFGqt74%3D&md5=17004209497660fb5f5cdda28a003d7dCAS |

Ayres, H., Martins, C. M., Ferreira, R. M., Mello, J. E., Dominguez, J. H., Souza, A. H., Valentin, R., Santos, I. C., and Baruselli, P. S. (2008). Effect of timing of estradiol benzoate administration upon synchronization of ovulation in suckling Nelore cows (Bos indicus) treated with a progesterone-releasing intravaginal device. Anim. Reprod. Sci. 109, 77–87.
Effect of timing of estradiol benzoate administration upon synchronization of ovulation in suckling Nelore cows (Bos indicus) treated with a progesterone-releasing intravaginal device.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXht12gu73J&md5=c9e49a73c6e99877743671ddba2a8428CAS |

Bao, B., Garverick, H. A., Smith, G. W., Smith, M. F., Salfen, B. E., and Youngquist, R. S. (1997). Changes in messenger ribonucleic acid encoding luteinizing hormone receptor, cytochrome P450-side chain cleavage, and aromatase are associated with recruitment and selection of bovine ovarian follicles. Biol. Reprod. 56, 1158–1168.
Changes in messenger ribonucleic acid encoding luteinizing hormone receptor, cytochrome P450-side chain cleavage, and aromatase are associated with recruitment and selection of bovine ovarian follicles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXisl2qs7w%3D&md5=5917a7938983a9018c42a852c2662574CAS |

Barcelos, A. C. Z., Satrapa, R. A., Nogueira, M. F. G., and Barros, C. M. (2006). Protocolo superestimulatório P-36 na raça Bonsmara: uso de eCG e atraso na indução da ovulação com LH. Acta Scientiae Veterinariae. 34, 514.

Barcelos, A. C. Z., Gouvêa, L. M., Meneghel, M., Barcelos, D. S., Barcelos, L. N., Trinca, L. A., and Barros, C. M. (2007). Efeito benéfico da substituição das duas últimas doses de pFSH por eCG no protocolo superestimulatório P-36, em vacas Nelore. Acta Scientiae Veterinariae. 35, 1260.

Barros, C. M., and Nogueira, M. F. (2001). Embryo transfer in Bos indicus cattle. Theriogenology 56, 1483–1496.
Embryo transfer in Bos indicus cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xjslyqtw%3D%3D&md5=3c9a7f888470d021b8c827b41d152feeCAS |

Barros, C. M., Moreira, M. B., Figueiredo, R. A., Teixeira, A. B., and Trinca, L. A. (2000). Synchronization of ovulation in beef cows (Bos indicus) using GnRH, PGF2alpha and estradiol benzoate. Theriogenology 53, 1121–1134.
Synchronization of ovulation in beef cows (Bos indicus) using GnRH, PGF2alpha and estradiol benzoate.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXjtl2jtbw%3D&md5=85eddedd0f580c3b2b7dd0720ecac92dCAS |

Barros, C. M., and Nogueira, M. F. G. (2005). Superovulation in zebu cattle: protocol P-36. Embryo Transfer Newsletter 23, 5–9.

Barros, C. M., Ereno, R. L., Simoes, R. A., Fernandes, P., Buratini, J., and Nogueira, M. F. (2010). Use of knowledge regarding LH receptors to improve superstimulatory treatments in cattle. Reprod. Fertil. Dev. 22, 132–137.
Use of knowledge regarding LH receptors to improve superstimulatory treatments in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXitlagu7s%3D&md5=ff1559c7fce8ef638710928983882762CAS |

Baruselli, P. S., de Sa Filho, M. F., Martins, C. M., Nasser, L. F., Nogueira, M. F., Barros, C. M., and Bo, G. A. (2006). Superovulation and embryo transfer in Bos indicus cattle. Theriogenology 65, 77–88.
Superovulation and embryo transfer in Bos indicus cattle.Crossref | GoogleScholarGoogle Scholar |

Beg, M. A., Bergfelt, D. R., Kot, K., Wiltbank, M. C., and Ginther, O. J. (2001). Follicular-fluid factors and granulosa-cell gene expression associated with follicle deviation in cattle. Biol. Reprod. 64, 432–441.
Follicular-fluid factors and granulosa-cell gene expression associated with follicle deviation in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXnsVKrsg%3D%3D&md5=390adc45fd87cefe1270e65765f0da35CAS |

Berfelt, D. R., Lightfoot, K. C., and Adams, G. P. (1994). Ovarian synchronization following ultrasound-guided transvaginal follicle ablation in heifers. Theriogenology 42, 895–907.
Ovarian synchronization following ultrasound-guided transvaginal follicle ablation in heifers.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD28zgtVemsA%3D%3D&md5=6ccde56232743d253a242cda6b3f663dCAS |

Bó, G. A., Baruselli, P. S., and Martínez, M. F. (2003). Pattern and manipulation of follicular development in Bos indicus cattle. Anim. Reprod. Sci. 78, 307–326.
Pattern and manipulation of follicular development in Bos indicus cattle.Crossref | GoogleScholarGoogle Scholar |

Bó, G. A., Baruselli, P. S., Chesta, P. M., and Martins, C. M. (2006). The timing of ovulation and insemination schedules in superstimulated cattle. Theriogenology 65, 89–101.
The timing of ovulation and insemination schedules in superstimulated cattle.Crossref | GoogleScholarGoogle Scholar |

Bó, G. A., Guerrero, D. C., and Adams, G. P. (2008). Alternative approaches to setting up donor cows for superstimulation. Theriogenology 69, 81–87.
Alternative approaches to setting up donor cows for superstimulation.Crossref | GoogleScholarGoogle Scholar |

Bodensteiner, K. J., Kot, K., Wiltbank, M. C., and Ginther, O. J. (1996). Synchronization of emergence of follicular waves in cattle. Theriogenology 45, 1115–1128.
Synchronization of emergence of follicular waves in cattle.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD28zgtVKhtA%3D%3D&md5=1593835d11044ce1ed4ea3f1afd4b6bcCAS |

Bucher, A., Kasimanickam, R., Hall, J. B., Dejarnette, J. M., Whittier, W. D., Kahn, W., and Xu, Z. (2009). Fixed-time AI pregnancy rate following insemination with frozen–thawed or fresh–extended semen in progesterone supplemented CO-Synch protocol in beef cows. Theriogenology 71, 1180–1185.
Fixed-time AI pregnancy rate following insemination with frozen–thawed or fresh–extended semen in progesterone supplemented CO-Synch protocol in beef cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXjsV2qs7o%3D&md5=e8866cc2ead47e5fa27b526143feb92aCAS |

Caixeta, E. S., Ripamonte, P., Franco, M. M., Junior, J. B., and Dode, M. A. (2009). Effect of follicle size on mRNA expression in cumulus cells and oocytes of Bos indicus: an approach to identify marker genes for developmental competence. Reprod. Fertil. Dev. 21, 655–664.
Effect of follicle size on mRNA expression in cumulus cells and oocytes of Bos indicus: an approach to identify marker genes for developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXmsVCrtbc%3D&md5=128f62c958507a48a575037ed9d74696CAS |

Callesen, H., Greve, T., and Hyttel, P. (1987). Premature ovulations in superovulated cattle. Theriogenology 28, 155–166.
Premature ovulations in superovulated cattle.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD283pvFSltQ%3D%3D&md5=7020bb6b200b68e657ac406ca4b47b1cCAS |

Carvalho, J. B., Carvalho, N. A., Reis, E. L., Nichi, M., Souza, A. H., and Baruselli, P. S. (2008). Effect of early luteolysis in progesterone-based timed AI protocols in Bos indicus, Bos indicus × Bos taurus, and Bos taurus heifers. Theriogenology 69, 167–175.
Effect of early luteolysis in progesterone-based timed AI protocols in Bos indicus, Bos indicus × Bos taurus, and Bos taurus heifers.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsVykug%3D%3D&md5=105c3c8ac7317a501cee48323e4286dfCAS |

Chesta, P., Tribulo, R., Tribulo, H., Balla, E., Baruselli, P. S., and Bó, G. A. (2007). Effect of the time of ovulation induction by gonadotropin-releasing hormone or pituitary luteinizing hormone on ova/embryo production in superstimulated beef cows inseminated at a fixed time. Reprod. Fertil. Dev. 19, 307.
Effect of the time of ovulation induction by gonadotropin-releasing hormone or pituitary luteinizing hormone on ova/embryo production in superstimulated beef cows inseminated at a fixed time.Crossref | GoogleScholarGoogle Scholar |

Conti, M., Hsieh, M., Park, J. Y., and Su, Y. Q. (2006). Role of the epidermal growth factor network in ovarian follicles. Mol. Endocrinol. 20, 715–723.
Role of the epidermal growth factor network in ovarian follicles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xkt1Oqu78%3D&md5=f36eac7c48afc829a0d6d51d90a3d6e2CAS |

D’Occhio, M. J., Sudha, G., Jillella, D., Whyte, T., Maclellan, L. J., Walsh, J., Trigg, T. E., and Miller, D. (1997). Use of a GnRH agonist to prevent the endogenous LH surge and injection of exogenous LH to induce ovulation in heifers superstimulated with FSH: a new model for superovulation. Theriogenology 47, 601–613.
Use of a GnRH agonist to prevent the endogenous LH surge and injection of exogenous LH to induce ovulation in heifers superstimulated with FSH: a new model for superovulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXhvFClt7c%3D&md5=3a1284a648287e75d3537fd59ebf1dc6CAS |

Dode, M. A., Dufort, I., Massicotte, L., and Sirard, M. A. (2006). Quantitative expression of candidate genes for developmental competence in bovine two-cell embryos. Mol. Reprod. Dev. 73, 288–297.
Quantitative expression of candidate genes for developmental competence in bovine two-cell embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhsVKht7o%3D&md5=f128c9356db4e0c65edbe5042bae2cf6CAS |

Donadeu, F. X., Esteves, C. L., Doyle, L. K., Walker, C. A., Schauer, S. N., and Diaz, C. A. (2011). Phospholipase Cbeta3 mediates LH-induced granulosa cell differentiation. Endocrinology 152, 2857–2869.
Phospholipase Cbeta3 mediates LH-induced granulosa cell differentiation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXosFaitLo%3D&md5=3b440c7ed9ef9ad338b856c7ffa90e0aCAS |

Donnison, M., and Pfeffer, P. L. (2004). Isolation of genes associated with developmentally competent bovine oocytes and quantitation of their levels during development. Biol. Reprod. 71, 1813–1821.
Isolation of genes associated with developmentally competent bovine oocytes and quantitation of their levels during development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXhtVWgsr%2FL&md5=e7fcfb720252293e1fc8b98090f99d63CAS |

Fair, T., Murphy, M., Rizos, D., Moss, C., Martin, F., Boland, M. P., and Lonergan, P. (2004). Analysis of differential maternal mRNA expression in developmentally competent and incompetent bovine two-cell embryos. Mol. Reprod. Dev. 67, 136–144.
Analysis of differential maternal mRNA expression in developmentally competent and incompetent bovine two-cell embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXjvVKkuw%3D%3D&md5=f2c8d806844fb647eecaa0d7bc71adb7CAS |

Fernandes, P., Teixeira, A. B., Crocci, A. J., and Barros, C. M. (2001). Timed artificial insemination in beef cattle using GnRH agonist, PGF2alpha and estradiol benzoate (EB). Theriogenology 55, 1521–1532.
Timed artificial insemination in beef cattle using GnRH agonist, PGF2alpha and estradiol benzoate (EB).Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXktVSns7c%3D&md5=eee8f0f800d95afc04551e56d32556dcCAS |

Ferreira, R., Oliveira, J. F., Fernandes, R., Moraes, J. F., and Goncalves, P. B. (2007). The role of angiotensin II in the early stages of bovine ovulation. Reproduction 134, 713–719.
The role of angiotensin II in the early stages of bovine ovulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXovVCiuw%3D%3D&md5=a7db1de303b303dbec5a3736869ad5d2CAS |

Fortune, J. E. (2001). Selection and maintenance of the dominant follicle: an introduction. Biol. Reprod. 65, 637.
| 1:CAS:528:DC%2BD3MXmtFenu7w%3D&md5=648b7ec5b96722aa114be0820c15b173CAS |

Fortune, J. E., Rivera, G. M., Evans, A. C., and Turzillo, A. M. (2001). Differentiation of dominant versus subordinate follicles in cattle. Biol. Reprod. 65, 648–654.
Differentiation of dominant versus subordinate follicles in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXmtFenu7o%3D&md5=72b8a42456e1b050b170063d2aee31f7CAS |

Gilchrist, R. B. (2011). Recent insights into oocyte–follicle cell interactions provide opportunities for the development of new approaches to in vitro maturation. Reprod. Fertil. Dev. 23, 23–31.
Recent insights into oocyte–follicle cell interactions provide opportunities for the development of new approaches to in vitro maturation.Crossref | GoogleScholarGoogle Scholar |

Gimenes, L. U., Sa Filho, M. F., Carvalho, N. A., Torres-Junior, J. R., Souza, A. H., Madureira, E. H., Trinca, L. A., Sartorelli, E. S., Barros, C. M., Carvalho, J. B., Mapletoft, R. J., and Baruselli, P. S. (2008). Follicle deviation and ovulatory capacity in Bos indicus heifers. Theriogenology 69, 852–858.
Follicle deviation and ovulatory capacity in Bos indicus heifers.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXktFCltr4%3D&md5=e1c96f6767572e3d6971b284069a3c4aCAS |

Ginther, O. J., Wiltbank, M. C., Fricke, P. M., Gibbons, J. R., and Kot, K. (1996). Selection of the dominant follicle in cattle. Biol. Reprod. 55, 1187–1194.
Selection of the dominant follicle in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXkvV2gtA%3D%3D&md5=2639fc239a504f12e284a15e2c856be0CAS |

Ginther, O. J., Beg, M. A., Bergfelt, D. R., Donadeu, F. X., and Kot, K. (2001). Follicle selection in monovular species. Biol. Reprod. 65, 638–647.
Follicle selection in monovular species.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXmtFenu70%3D&md5=0c33448b939e97b8adcba4057382c097CAS |

Ginther, O. J., Beg, M. A., Donadeu, F. X., and Bergfelt, D. R. (2003). Mechanism of follicle deviation in monovular farm species. Anim. Reprod. Sci. 78, 239–257.
Mechanism of follicle deviation in monovular farm species.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXksF2gtro%3D&md5=be23f866eecb92e907b79f9b2bc005ccCAS |

Goulding, D., Williams, D. H., Duffy, P., Boland, M. P., and Roche, J. F. (1990). Superovulation in heifers given FSH initiated either at Day 2 or Day 10 of the estrous cycle. Theriogenology 34, 767–778.
Superovulation in heifers given FSH initiated either at Day 2 or Day 10 of the estrous cycle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXksVaht7s%3D&md5=de66f700b3d9ddf383198acbb080e7dfCAS |

Guilbault, L. A., Grasso, F., Lussier, J. G., Rouillier, P., and Matton, P. (1991). Decreased superovulatory responses in heifers superovulated in the presence of a dominant follicle. J. Reprod. Fertil. 91, 81–89.
Decreased superovulatory responses in heifers superovulated in the presence of a dominant follicle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXisVSkur4%3D&md5=c82814e088f5ed05dbf4f05fd8da831eCAS |

Herrlich, A., Kuhn, B., Grosse, R., Schmid, A., Schultz, G., and Gudermann, T. (1996). Involvement of Gs and Gi proteins in dual coupling of the luteinizing hormone receptor to adenylyl cyclase and phospholipase C. J. Biol. Chem. 271, 16 764–16 772.
Involvement of Gs and Gi proteins in dual coupling of the luteinizing hormone receptor to adenylyl cyclase and phospholipase C.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28Xkt1CjsLs%3D&md5=0330c56d53b2b70c25d1de2394170ef4CAS |

Hosoe, M., Kaneyama, K., Ushizawa, K., Hayashi, K. G., and Takahashi, T. (2011). Quantitative analysis of bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) gene expression in calf and adult bovine ovaries. Reprod. Biol. Endocrinol. 9, 33.
Quantitative analysis of bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) gene expression in calf and adult bovine ovaries.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXjvFCjtLo%3D&md5=d4745d8d9d1f7656e497e19051512520CAS |

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 | 1:CAS:528:DC%2BD28XotV2gsb4%3D&md5=0f9b6f0fe43b776d7f08409072e0f775CAS |

Hyttel, P., Callesen, H., and Greve, T. (1986). Ultrastructural features of preovulatory oocyte maturation in superovulated cattle. J. Reprod. Fertil. 76, 645–656.
Ultrastructural features of preovulatory oocyte maturation in superovulated cattle.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL287pvFyrtQ%3D%3D&md5=fde3fe977ec1a4398f8221e63fb18148CAS |

Ji, I., and Ji, T. H. (1991). Exons 1–10 of the rat LH receptor encode a high affinity hormone binding site and exon 11 encodes G-protein modulation and a potential second hormone binding site. Endocrinology 128, 2648–2650.
Exons 1–10 of the rat LH receptor encode a high affinity hormone binding site and exon 11 encodes G-protein modulation and a potential second hormone binding site.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXksVeitr0%3D&md5=b852c729ba593c584341cf9d9ef80471CAS |

Ji, T. H., Grossmann, M., and Ji, I. (1998). G protein-coupled receptors. I. Diversity of receptor–ligand interactions. J. Biol. Chem. 273, 17 299–17 302.
G protein-coupled receptors. I. Diversity of receptor–ligand interactions.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXkvFaktbo%3D&md5=a899236cf7a79bba1149b73c8dc6883eCAS |

Kash, J. C., and Menon, K. M. (1998). Identification of a hormonally regulated luteinizing hormone/human chorionic gonadotropin receptor mRNA binding protein. Increased mrna binding during receptor down-regulation. J. Biol. Chem. 273, 10 658–10 664.
Identification of a hormonally regulated luteinizing hormone/human chorionic gonadotropin receptor mRNA binding protein. Increased mrna binding during receptor down-regulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXivFKnu7o%3D&md5=240965413b46e71af108e6b012722119CAS |

Kash, J. C., and Menon, K. M. (1999). Sequence-specific binding of a hormonally regulated mRNA binding protein to cytidine-rich sequences in the lutropin receptor open reading frame. Biochemistry 38, 16 889–16 897.
Sequence-specific binding of a hormonally regulated mRNA binding protein to cytidine-rich sequences in the lutropin receptor open reading frame.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXnsFyisLc%3D&md5=1def9355ba6823b8a08f6ef34fa86572CAS |

Killian, G. J. (2004). Evidence for the role of oviduct secretions in sperm function, fertilization and embryo development. Anim. Reprod. Sci. 82-83, 141–153.
Evidence for the role of oviduct secretions in sperm function, fertilization and embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXmtVCit7w%3D&md5=019e0b337065c82d85a98c820e842da6CAS |

Koo, Y. B., Ji, I., Slaughter, R. G., and Ji, T. H. (1991). Structure of the luteinizing hormone receptor gene and multiple exons of the coding sequence. Endocrinology 128, 2297–2308.
Structure of the luteinizing hormone receptor gene and multiple exons of the coding sequence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK38XhsFWk&md5=31c2c44e59d8aefd84fb734d5b74acbaCAS |

Kuji, N., Sueoka, K., Miyazaki, T., Tanaka, M., Oda, T., Kobayashi, T., and Yoshimura, Y. (1996). Involvement of angiotensin II in the process of gonadotropin-induced ovulation in rabbits. Biol. Reprod. 55, 984–991.
Involvement of angiotensin II in the process of gonadotropin-induced ovulation in rabbits.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28Xmtlamt7g%3D&md5=24ef6130e106741a1bae206d70ed48bcCAS |

Larson, J. E., Thielen, K. N., Funnell, B. J., Stevenson, J. S., Kesler, D. J., and Lamb, G. C. (2009). Influence of a controlled internal drug release after fixed-time artificial insemination on pregnancy rates and returns to estrus of nonpregnant cows. J. Anim. Sci. 87, 914–921.
Influence of a controlled internal drug release after fixed-time artificial insemination on pregnancy rates and returns to estrus of nonpregnant cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXislarurg%3D&md5=e08864f788cc341951b3b94653e33d94CAS |

Lei, Z. M., Reshef, E., and Rao, V. (1992). The expression of human chorionic gonadotropin/luteinizing hormone receptors in human endometrial and myometrial blood vessels. J. Clin. Endocrinol. Metab. 75, 651–659.
The expression of human chorionic gonadotropin/luteinizing hormone receptors in human endometrial and myometrial blood vessels.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK38Xls1Chs74%3D&md5=b68291a1bd58bafa3454dc8f60afd785CAS |

Lerner, S. P., Thayne, W. V., Baker, R. D., Henschen, T., Meredith, S., Inskeep, E. K., Dailey, R. A., Lewis, P. E., and Butcher, R. L. (1986). Age, dose of FSH and other factors affecting superovulation in Holstein cows. J. Anim. Sci. 63, 176–183.
| 1:CAS:528:DyaL28XksFCktrc%3D&md5=556ac48911365bccb427cf4c76c21321CAS |

Liu, J., and Sirois, J. (1998). Follicle size-dependent induction of prostaglandin G/H synthase-2 during superovulation in cattle. Biol. Reprod. 58, 1527–1532.
Follicle size-dependent induction of prostaglandin G/H synthase-2 during superovulation in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXjsFShur4%3D&md5=4b3e794f95b5dbfc6ac9c2cbbb6b77ccCAS |

Looney, C. R., and Pryor, J. H. (2010). Practical applications of new research information in the practice of bovine embryo transfer. Reprod. Fertil. Dev. 22, 145–150.
Practical applications of new research information in the practice of bovine embryo transfer.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1MfitlGiug%3D%3D&md5=55726971e4d6e953018818a237a310e1CAS |

Lucidi, P., Bernabo, N., Turriani, M., Barboni, B., and Mattioli, M. (2003). Cumulus cells steroidogenesis is influenced by the degree of oocyte maturation. Reprod. Biol. Endocrinol. 1, 45.
Cumulus cells steroidogenesis is influenced by the degree of oocyte maturation.Crossref | GoogleScholarGoogle Scholar |

Mapletoft, R. J., and Hasler, J. F. (2005). Assisted reproductive technologies in cattle: a review. Rev. Sci. Tech. 24, 393–403.
| 1:STN:280:DC%2BD2MvksVaitw%3D%3D&md5=319246670cbb281330b43a3d5cf9ef66CAS |

Mapletoft, R. J., Steward, K. B., and Adams, G. P. (2002). Recent advances in the superovulation in cattle. Reprod. Nutr. Dev. 42, 601–611.
Recent advances in the superovulation in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXit1ymtr8%3D&md5=18b09d4c6bb19af7006a027e83de8573CAS |

Martinez, M. F., Kastelic, J. P., Adams, G. P., and Mapletoft, R. J. (2002). The use of a progesterone-releasing device (CIDR-B) or melengestrol acetate with GnRH, LH, or estradiol benzoate for fixed-time AI in beef heifers. J. Anim. Sci. 80, 1746–1751.
| 1:CAS:528:DC%2BD38XmvVemu7k%3D&md5=2668c9351f9eee75fed8d1050ec8eb8fCAS |

Mattos, M. C., Bastos, M. R., Guardieiro, M. M., Carvalho, J. O., Franco, M. M., Mourao, G. B., Barros, C. M., and Sartori, R. (2011). Improvement of embryo production by the replacement of the last two doses of porcine follicle-stimulating hormone with equine chorionic gonadotropin in Sindhi donors. Anim. Reprod. Sci. 125, 119–123.
Improvement of embryo production by the replacement of the last two doses of porcine follicle-stimulating hormone with equine chorionic gonadotropin in Sindhi donors.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXnsVyrtb8%3D&md5=448538fc62faa8b6ec5ddae1d87b279aCAS |

Meneghetti, M., Sa Filho, O. G., Peres, R. F., Lamb, G. C., and Vasconcelos, J. L. (2009). Fixed-time artificial insemination with estradiol and progesterone for Bos indicus cows I: basis for development of protocols. Theriogenology 72, 179–189.
Fixed-time artificial insemination with estradiol and progesterone for Bos indicus cows I: basis for development of protocols.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXnsFSjtro%3D&md5=55ef4dddefe192f3adf07959c673d836CAS |

Menon, B., Peegel, H., and Menon, K. M. (2009). Evidence for the association of luteinizing hormone receptor mRNA-binding protein with the translating ribosomes during receptor downregulation. Biochim. Biophys. Acta 1793, 1787–1794.
Evidence for the association of luteinizing hormone receptor mRNA-binding protein with the translating ribosomes during receptor downregulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXht1yjtbrJ&md5=1164893313daf658963d5354489fa719CAS |

Mourot, M., Dufort, I., Gravel, C., Algriany, O., Dieleman, S., and Sirard, M. A. (2006). The influence of follicle size, FSH-enriched maturation medium, and early cleavage on bovine oocyte maternal mRNA levels. Mol. Reprod. Dev. 73, 1367–1379.
The influence of follicle size, FSH-enriched maturation medium, and early cleavage on bovine oocyte maternal mRNA levels.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtVSksb7K&md5=c08acc3238df0800f26f269236c05e6bCAS |

Murphy, B. D., and Martinuk, S. D. (1991). Equine chorionic gonadotropin. Endocr. Rev. 12, 27–44.
Equine chorionic gonadotropin.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXlt1Oiu7k%3D&md5=456531f3ba1168c8362f7817afe8ff9eCAS |

Nair, A. K., Kash, J. C., Peegel, H., and Menon, K. M. (2002). Post-transcriptional regulation of luteinizing hormone receptor mRNA in the ovary by a novel mRNA-binding protein. J. Biol. Chem. 277, 21 468–21 473.
Post-transcriptional regulation of luteinizing hormone receptor mRNA in the ovary by a novel mRNA-binding protein.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xkslyqtbw%3D&md5=583cbde4b825dbbd8926ae291a32ebe6CAS |

Nasser, L. F., Adams, G. P., Bo, G. A., and Mapletoft, R. J. (1993). Ovarian superstimulatory response relative to follicular wave emergence in heifers. Theriogenology 40, 713–724.
Ovarian superstimulatory response relative to follicular wave emergence in heifers.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD28zgtVWitg%3D%3D&md5=6aa4723e4ad1abf484eb6f0ae21761fcCAS |

Nasser, L. F., Sa Filho, M. F., Reis, E. L., Rezende, C. R., Mapletoft, R. J., Bo, G. A., and Baruselli, P. S. (2011). Exogenous progesterone enhances ova and embryo quality following superstimulation of the first follicular wave in Nelore (Bos indicus) donors. Theriogenology 76, 320–327.
Exogenous progesterone enhances ova and embryo quality following superstimulation of the first follicular wave in Nelore (Bos indicus) donors.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXnsFWiur4%3D&md5=45216c29d48f3466f4c86473e8a038e8CAS |

Nogueira, M. F. G., and Barros, C. M. (2003). Timing of ovulation in Nelore cows superstimulated with P36 protocol. Acta Scientiae Veterinariae 31, 509.

Nogueira, M. F., Melo, D. S., Carvalho, L. M., Fuck, E. J., Trinca, L. A., and Barros, C. M. (2004). Do high progesterone concentrations decrease pregnancy rates in embryo recipients synchronized with PGF2alpha and eCG? Theriogenology 61, 1283–1290.
Do high progesterone concentrations decrease pregnancy rates in embryo recipients synchronized with PGF2alpha and eCG?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXhsVOmtbg%3D&md5=a0b2f2d6c048bf2d0e847ee56577ca1fCAS |

Nogueira, M. F., Buratini, J., Price, C. A., Castilho, A. C., Pinto, M. G., and Barros, C. M. (2007a). Expression of LH receptor mRNA splice variants in bovine granulosa cells: changes with follicle size and regulation by FSH in vitro. Mol. Reprod. Dev. 74, 680–686.
Expression of LH receptor mRNA splice variants in bovine granulosa cells: changes with follicle size and regulation by FSH in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXltVGrtro%3D&md5=d753160a58b491e94c13d169148f75dcCAS |

Nogueira, M. F., Fragnito, P. S., Trinca, L. A., and Barros, C. M. (2007b). The effect of type of vaginal insert and dose of pLH on embryo production, following fixed-time AI in a progestin-based superstimulatory protocol in Nelore cattle. Theriogenology 67, 655–660.
The effect of type of vaginal insert and dose of pLH on embryo production, following fixed-time AI in a progestin-based superstimulatory protocol in Nelore cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXitl2jtw%3D%3D&md5=41fce965079431b028ce27bfa86fed4aCAS |

Oliveira, A. C. S., Mattos, M. C. C., Bastos, M. R., Lunardi, L. H., Surjus, R. S., Sartori, R., and Barros, C. M. (2010). Eficiência do protocolo superestimulatório P-36, associado à administração de eCG ou LH, em vacas da raça Nelore. Acta Scientiae Veterinariae 38, 352.

Pangas, S. A., and Matzuk, M. M. (2005). The art and artifact of GDF9 activity: cumulus expansion and the cumulus expansion-enabling factor. Biol. Reprod. 73, 582–585.
The art and artifact of GDF9 activity: cumulus expansion and the cumulus expansion-enabling factor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtVCgtrrP&md5=bbb38e2f97407b0cbe54ddb82d4644e8CAS |

Panigone, S., Hsieh, M., Fu, M., Persani, L., and Conti, M. (2008). Luteinizing hormone signaling in preovulatory follicles involves early activation of the epidermal growth factor receptor pathway. Mol. Endocrinol. 22, 924–936.
Luteinizing hormone signaling in preovulatory follicles involves early activation of the epidermal growth factor receptor pathway.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXkt1Cnsbs%3D&md5=20a350fd1dbc582bdd0a447d0c2b288cCAS |

Paul, M., Poyan Mehr, A., and Kreutz, R. (2006). Physiology of local renin–angiotensin systems. Physiol. Rev. 86, 747–803.
Physiology of local renin–angiotensin systems.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XnsFGgtLk%3D&md5=4412382d04289c9b9c33d8b1181392cfCAS |

Pellicer, A., Palumbo, A., DeCherney, A. H., and Naftolin, F. (1988). Blockage of ovulation by an angiotensin antagonist. Science 240, 1660–1661.
Blockage of ovulation by an angiotensin antagonist.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1cXktlaht7w%3D&md5=dbd98a6ea5ebb2b228499bd8a6d9ef4dCAS |

Peng, X. R., Hsueh, A. J., LaPolt, P. S., Bjersing, L., and Ny, T. (1991). Localization of luteinizing hormone receptor messenger ribonucleic acid expression in ovarian cell types during follicle development and ovulation. Endocrinology 129, 3200–3207.
Localization of luteinizing hormone receptor messenger ribonucleic acid expression in ovarian cell types during follicle development and ovulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK38XjsVyhtg%3D%3D&md5=89c39035964eed717176eb2134e94e37CAS |

Perry, G. A., Smith, M. F., and Patterson, D. J. (2002). Evaluation of a fixed-time artificial insemination protocol for postpartum suckled beef cows. J. Anim. Sci. 80, 3060–3064.
| 1:CAS:528:DC%2BD3sXhvFSisQ%3D%3D&md5=7e2f10da6b5ca9b54ffab0c69f4da8bcCAS |

Pinheiro, O. L., Barros, C. M., Figueiredo, R. A., do Valle, E. R., Encarnacao, R. O., and Padovani, C. R. (1998). Estrous behavior and the estrus-to-ovulation interval in Nelore cattle (Bos indicus) with natural estrus or estrus induced with prostaglandin F2 alpha or norgestomet and estradiol valerate. Theriogenology 49, 667–681.
Estrous behavior and the estrus-to-ovulation interval in Nelore cattle (Bos indicus) with natural estrus or estrus induced with prostaglandin F2 alpha or norgestomet and estradiol valerate.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXhtFShu7Y%3D&md5=a371cfb6a4c538aa0fb846a010cbc3dcCAS |

Pinheiro, V. G., Souza, A. F., Pegorer, M. F., Satrapa, R. A., Ereno, R. L., Trinca, L. A., and Barros, C. M. (2009). Effects of temporary calf removal and eCG on pregnancy rates to timed-insemination in progesterone-treated postpartum Nellore cows. Theriogenology 71, 519–524.
Effects of temporary calf removal and eCG on pregnancy rates to timed-insemination in progesterone-treated postpartum Nellore cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsFChtLzK&md5=889792e89335b7328d5e2bd130816f27CAS |

Portela, V. M., Goncalves, P. B., Veiga, A. M., Nicola, E., Buratini, J., and Price, C. A. (2008). Regulation of angiotensin type 2 receptor in bovine granulosa cells. Endocrinology 149, 5004–5011.
Regulation of angiotensin type 2 receptor in bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtF2gtbbK&md5=852c17bbb12f03273863666052c500f5CAS |

Pursley, J. R., Mee, M. O., and Wiltbank, M. C. (1995). Synchronization of ovulation in dairy cows using PGF2alpha and GnRH. Theriogenology 44, 915–923.
Synchronization of ovulation in dairy cows using PGF2alpha and GnRH.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXpvFymu7w%3D&md5=adc07b2b8f1c1e7b8b39ef736240b80fCAS |

Reano, I., Carballo, D., Tribulo, A., Tribulo, P., Balla, E., and Bo, G. A. (2009). Efecto de la adicion de eCG a los tratamientos superovulatorios con Folltropin-V en la produccion de embriones de donantes de embriones. VIII Simposio Internacional de Reproduccion animal – IRAC 1, 54.

Reizel, Y., Elbaz, J., and Dekel, N. (2010). Sustained activity of the EGF receptor is an absolute requisite for LH-induced oocyte maturation and cumulus expansion. Mol. Endocrinol. 24, 402–411.
Sustained activity of the EGF receptor is an absolute requisite for LH-induced oocyte maturation and cumulus expansion.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhslejsrs%3D&md5=754d1277c454534d8304d3409dcde375CAS |

Roberts, A. J., Grizzle, J. M., and Echternkamp, S. E. (1994). Follicular development and superovulation response in cows administered multiple FSH injections early in the estrous cycle. Theriogenology 42, 917–929.
Follicular development and superovulation response in cows administered multiple FSH injections early in the estrous cycle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXis1Smsrk%3D&md5=dd0a7767220b2472566d9e010e357e08CAS |

Rosa, F. S., Bonotto, A. L. M., Trinca, L. A., Nogueira, M. G. F., and Barros, C. M. (2010). Eficiência do protocolo superestimulatório P-36, associado a administração de eCG ou LH em doadoras da raça Angus. (resumo). Acta Scientiae Veterinariae 37, 711.

Sa Filho, O. G., Meneghetti, M., Peres, R. F., Lamb, G. C., and Vasconcelos, J. L. (2009). Fixed-time artificial insemination with estradiol and progesterone for Bos indicus cows II: strategies and factors affecting fertility. Theriogenology 72, 210–218.
Fixed-time artificial insemination with estradiol and progesterone for Bos indicus cows II: strategies and factors affecting fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXnsFSjtrs%3D&md5=809d9574d43f619c4ab0c48c1528a31cCAS |

Sartorelli, E. S., Carvalho, L. M., Bergfelt, D. R., Ginther, O. J., and Barros, C. M. (2005). Morphological characterization of follicle deviation in Nelore (Bos indicus) heifers and cows. Theriogenology 63, 2382–2394.
Morphological characterization of follicle deviation in Nelore (Bos indicus) heifers and cows.Crossref | GoogleScholarGoogle Scholar |

Sartori, R., and Barros, C. M. (2011). Reproductive cycles in Bos indicus cattle. Anim. Reprod. Sci. 124, 244–250.
Reproductive cycles in Bos indicus cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXlvFWhur4%3D&md5=446e7abbd84e5ef33439a6fb85b404caCAS |

Sartori, R., Fricke, P. M., Ferreira, J. C., Ginther, O. J., and Wiltbank, M. C. (2001). Follicular deviation and acquisition of ovulatory capacity in bovine follicles. Biol. Reprod. 65, 1403–1409.
Follicular deviation and acquisition of ovulatory capacity in bovine follicles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXnvVersrk%3D&md5=d6870866bb1663618bb837c21152c4d1CAS |

Shemesh, M. (2001). Actions of gonadotrophins on the uterus. Reproduction 121, 835–842.
Actions of gonadotrophins on the uterus.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXkslWnsbk%3D&md5=45263b5debdb98fd08fbb2e98a382d25CAS |

Shi, H., and Segaloff, D. L. (1995). A role for increased lutropin/choriogonadotropin receptor (LHR) gene transcription in the follitropin-stimulated induction of the LHR in granulosa cells. Mol. Endocrinol. 9, 734–744.
A role for increased lutropin/choriogonadotropin receptor (LHR) gene transcription in the follitropin-stimulated induction of the LHR in granulosa cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXmtVCrt7s%3D&md5=af250efed63ed3575742883b8f5803caCAS |

Simões, R. A., Satrapa, R. A., Rosa, F. S., Piagentini, M., Castilho, A. C., Ereno, R. L., Trinca, L. A., Nogueira, M. F., Buratini, J., and Barros, C. M. (2012). Ovulation rate and its relationship with follicle diameter and gene expression of the LH receptor (LHR) in Nelore cows. Theriogenology 77, 139–147.
Ovulation rate and its relationship with follicle diameter and gene expression of the LH receptor (LHR) in Nelore cows.Crossref | GoogleScholarGoogle Scholar |

Sirard, M. A., Richard, F., Blondin, P., and Robert, C. (2006). Contribution of the oocyte to embryo quality. Theriogenology 65, 126–136.
Contribution of the oocyte to embryo quality.Crossref | GoogleScholarGoogle Scholar |

Sirard, M. A. (2007). From biological fundamentals to practice, and back. Theriogenology 68, S250–S256.
From biological fundamentals to practice, and back.Crossref | GoogleScholarGoogle Scholar |

Stock, A. E., Ellington, J. E., and Fortune, J. E. (1996). A dominant follicle does not affect follicular recruitment by superovulatory doses of FSH in cattle but can inhibit ovulation. Theriogenology 45, 1091–1102.
A dominant follicle does not affect follicular recruitment by superovulatory doses of FSH in cattle but can inhibit ovulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XjtVWrtL0%3D&md5=45c5fc76f1b4ef37c65381e952c2c6c5CAS |

Tsai-Morris, C. H., Buczko, E., Wang, W., Xie, X. Z., and Dufau, M. L. (1991). Structural organization of the rat luteinizing hormone (LH) receptor gene. J. Biol. Chem. 266, 11 355–11 359.
| 1:CAS:528:DyaK38XhsVGju74%3D&md5=719af83266c3357b1b3cece4700a2f47CAS |

van de Leemput, E. E., Vos, P. L., Hyttel, P., van den Hurk, R., Bevers, M. M., van der Weijden, G. C., and Dieleman, S. J. (2001). Effects of brief postponement of the preovulatory LH surge on ovulation rates and embryo formation in eCG/prostaglandin-treated heifers. Theriogenology 55, 573–592.
Effects of brief postponement of the preovulatory LH surge on ovulation rates and embryo formation in eCG/prostaglandin-treated heifers.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXhsFCqu7s%3D&md5=4cf42f6c497c8fb6bbadab1bfae0639aCAS |

Xu, Z., Garverick, H. A., Smith, G. W., Smith, M. F., Hamilton, S. A., and Youngquist, R. S. (1995). Expression of messenger ribonucleic acid encoding cytochrome P450 side-chain cleavage, cytochrome p450 17 alpha-hydroxylase, and cytochrome P450 aromatase in bovine follicles during the first follicular wave. Endocrinology 136, 981–989.
Expression of messenger ribonucleic acid encoding cytochrome P450 side-chain cleavage, cytochrome p450 17 alpha-hydroxylase, and cytochrome P450 aromatase in bovine follicles during the first follicular wave.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXjvFCjsbo%3D&md5=a17188e37593fcb29da738aa07408b46CAS |

Yoshimura, Y., Karube, M., Oda, T., Koyama, N., Shiokawa, S., Akiba, M., Yoshinaga, A., and Nakamura, Y. (1993). Locally produced angiotensin II induces ovulation by stimulating prostaglandin production in in vitro perfused rabbit ovaries. Endocrinology 133, 1609–1616.
Locally produced angiotensin II induces ovulation by stimulating prostaglandin production in in vitro perfused rabbit ovaries.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2cXhtlyg&md5=436ac734087592898de1d1956511fddaCAS |

Yoshimura, Y., Karube, M., Aoki, H., Oda, T., Koyama, N., Nagai, A., Akimoto, Y., Hirano, H., and Nakamura, Y. (1996). Angiotensin II induces ovulation and oocyte maturation in rabbit ovaries via the AT2 receptor subtype. Endocrinology 137, 1204–1211.
Angiotensin II induces ovulation and oocyte maturation in rabbit ovaries via the AT2 receptor subtype.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XhvVegsbo%3D&md5=ed69f63c9f37b6e3d41604208631a690CAS |

Zhang, M., Shi, H., Segaloff, D. L., and Van Voorhis, B. J. (2001). Expression and localization of luteinizing hormone receptor in the female mouse reproductive tract. Biol. Reprod. 64, 179–187.
| 1:STN:280:DC%2BD3M7gsl2rsw%3D%3D&md5=621487970a8f8f2b17d44e2dbbbeddf9CAS |