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

Calcium-sensing receptor (CASR) is involved in porcine in vitro fertilisation and early embryo development

C. Liu A , Y. Liu B , K. Larsen C , Y. P. Hou A and H. Callesen B D
+ Author Affiliations
- Author Affiliations

A State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China.

B Department of Animal Science, Aarhus University, Blichers Allé 20, DK-8830 Tjele, Denmark.

C Department of Molecular Biology and Genetics, Aarhus University, Blichers Allé 20, DK-8830 Tjele, Denmark.

D Corresponding author. Email: henrik.callesen@anis.au.dk

Reproduction, Fertility and Development 30(2) 391-398 https://doi.org/10.1071/RD16338
Submitted: 27 August 2016  Accepted: 25 June 2017   Published: 17 July 2017

Abstract

It has been demonstrated that extracellular calcium is necessary in fertilisation and embryo development but the mechanism is still not well understood. The present study mainly focussed on the extracellular calcium effector called the calcium-sensing receptor (CASR) and examined its expression in porcine gametes and embryos and its function during fertilisation and early embryo development. By using reverse transcription polymerase chain reaction, CASR was found to be expressed in porcine oocytes, spermatozoa and embryos at different developmental stages. Functionally, medium supplementation with a CASR agonist or an antagonist during in vitro fertilisation (IVF) and in vitro culture (IVC) was tested. During fertilisation, the presence of a CASR agonist increased sperm penetration rate and decreased polyspermy rate leading to an increased normal fertilisation rate. During embryo development, for the IVF embryos, agonist treatment during IVC significantly increased cleavage rate and blastocyst formation rate compared with the control group. Furthermore, parthenogenetically activated embryos showed similar results with lower cleavage and blastocyst formation rates in the antagonist group than in the other groups. It was concluded that CASR, as the effector of extracellular calcium, modulates porcine fertilisation and early embryo development.

Additional keywords: agonist, antagonist, parthenogenetic activation, pig.


References

Abdoon, A. S., Ghanem, N., Kandil, O. M., Gad, A., Schellander, K., and Tesfaye, D. (2012). cDNA microarray analysis of gene expression in parthenotes and in vitro produced buffalo embryos. Theriogenology 77, 1240–1251.
cDNA microarray analysis of gene expression in parthenotes and in vitro produced buffalo embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xjs1amurk%3D&md5=63cdb783cbc42d8d12fa7b6a98eb1cfeCAS |

Avlani, V. A., Ma, W., Mun, H. C., Leach, K., Delbridge, L., Christopoulos, A., and Conigrave, A. D. (2013). Calcium-sensing receptor-dependent activation of CREB phosphorylation in HEK293 cells and human parathyroid cells. Am. J. Physiol. Endocrinol. Metab. 304, E1097–E1104.
Calcium-sensing receptor-dependent activation of CREB phosphorylation in HEK293 cells and human parathyroid cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXpt1yjtL0%3D&md5=07f9dab62bc08ef5c750edfe9683a558CAS |

Beek, J., Maes, D., Nauwynck, H., Piepers, S., and Van Soom, A. (2015a). A critical assessment of the effect of serine protease inhibitors on porcine fertilization and quality parameters of porcine spermatozoa in vitro. Reprod. Biol. 15, 9–19.
A critical assessment of the effect of serine protease inhibitors on porcine fertilization and quality parameters of porcine spermatozoa in vitro.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC2MngtVGhtQ%3D%3D&md5=746339fa33348a0e2a37ed599cb96036CAS |

Beek, J., Nauwynck, H., Appeltant, R., Maes, D., and Van Soom, A. (2015b). Inhibitors of serine proteases decrease sperm penetration during porcine fertilization in vitro by inhibiting sperm binding to the zona pellucida and acrosome reaction. Theriogenology 84, 1378–1386.
Inhibitors of serine proteases decrease sperm penetration during porcine fertilization in vitro by inhibiting sperm binding to the zona pellucida and acrosome reaction.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXht12ku7vK&md5=1b0faf77ae3146c26aeb15f6e7b2b262CAS |

Boccaccio, A., Frassanito, M. C., Lamberti, L., Brunelli, R., Maulucci, G., Monaci, M., Papi, M., Pappalettere, C., Parasassi, T., Sylla, L., Ursini, F., and De Spirito, M. (2012). Nanoscale characterization of the biomechanical hardening of bovine zona pellucida. J. R. Soc. Interface 9, 2871–2882.
Nanoscale characterization of the biomechanical hardening of bovine zona pellucida.Crossref | GoogleScholarGoogle Scholar |

Colella, M., Gerbino, A., Hofer, A. M., and Curci, S. (2016). Recent advances in understanding the extracellular calcium-sensing receptor. F1000Res. 5, F1000 Faculty Rev-2535.
Recent advances in understanding the extracellular calcium-sensing receptor.Crossref | GoogleScholarGoogle Scholar |

Cowell, H. E., and Garrod, D. R. (1999). Activation of protein kinase C modulates cell–cell and cell–substratum adhesion of a human colorectal carcinoma cell line and restores ‘normal’ epithelial morphology. Int. J. Cancer 80, 455–464.
Activation of protein kinase C modulates cell–cell and cell–substratum adhesion of a human colorectal carcinoma cell line and restores ‘normal’ epithelial morphology.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXptVSqtw%3D%3D&md5=5a2b319f52d9154be327acb796edb39dCAS |

Dal Pra, I., Chiarini, A., Pacchiana, R., Gardenal, E., Chakravarthy, B., Whitfield, J. F., and Armato, U. (2014). Calcium-sensing receptors of human astrocyte–neuron teams: amyloid-beta-driven mediators and therapeutic targets of Alzheimer’s disease. Curr. Neuropharmacol. 12, 353–364.
Calcium-sensing receptors of human astrocyte–neuron teams: amyloid-beta-driven mediators and therapeutic targets of Alzheimer’s disease.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC2M3jtFSjtA%3D%3D&md5=035657c884346d771a088c28a823ce4cCAS |

Dell’Aquila, M. E., De Santis, T., Cho, Y. S., Reshkin, S. J., Caroli, A. M., Maritato, F., Minoia, P., and Casavola, V. (2006). Localization and quantitative expression of the calcium-sensing receptor protein in human oocytes. Fertil. Steril. 85, 1240–1247.
Localization and quantitative expression of the calcium-sensing receptor protein in human oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XksFWjsrw%3D&md5=b889f92107e245c68ee3beba0b6ce169CAS |

De Santis, T., Casavola, V., Reshkin, S. J., Guerra, L., Ambruosi, B., Fiandanese, N., Dalbies-Tran, R., Goudet, G., and Dell’Aquila, M. E. (2009). The extracellular calcium-sensing receptor is expressed in the cumulus–oocyte complex in mammals and modulates oocyte meiotic maturation. Reproduction 138, 439–452.
The extracellular calcium-sensing receptor is expressed in the cumulus–oocyte complex in mammals and modulates oocyte meiotic maturation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtFOgtrvN&md5=faec2582e076546dcdb9e1044d782e07CAS |

González-Vélez, V., and Godínez-Fernández, J. R. (2004). Simulation of five intracellular C2+-regulation mechanisms in response to voltage-clamp pulses. Comput. Biol. Med. 34, 279–292.
Simulation of five intracellular C2+-regulation mechanisms in response to voltage-clamp pulses.Crossref | GoogleScholarGoogle Scholar |

Hobson, S. A., Wright, J., Lee, F., McNeil, S. E., Bilderback, T., and Rodland, K. D. (2003). Activation of the MAP kinase cascade by exogenous calcium-sensing receptor. Mol. Cell. Endocrinol. 200, 189–198.
Activation of the MAP kinase cascade by exogenous calcium-sensing receptor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXitV2hsbk%3D&md5=24aced3673ba3e3a9f7522a34d83d1e5CAS |

Hofer, A. M., and Brown, E. M. (2003). Extracellular calcium sensing and signalling. Nat. Rev. Mol. Cell Biol. 4, 530–538.
Extracellular calcium sensing and signalling.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXltVWmsr4%3D&md5=374d15bf229f8ef7c5118c09cca19e5eCAS |

Huang, S., Maher, V. M., and McCormick, J. J. (1995). Extracellular Ca2+ stimulates the activation of mitogen-activated protein kinase and cell growth in human fibroblasts. Biochem. J. 310, 881–885.
Extracellular Ca2+ stimulates the activation of mitogen-activated protein kinase and cell growth in human fibroblasts.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXot1aktLc%3D&md5=b29b761fc4c96526e84f360ec75c0426CAS |

Kifor, O., Diaz, R., Butters, R., and Brown, E. M. (1997). The Ca2+-sensing receptor (CaR) activates phospholipases C, A2, and D in bovine parathyroid and CaR-transfected, human embryonic kidney (HEK293) cells. J. Bone Miner. Res. 12, 715–725.
The Ca2+-sensing receptor (CaR) activates phospholipases C, A2, and D in bovine parathyroid and CaR-transfected, human embryonic kidney (HEK293) cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXjtlOqtrw%3D&md5=6de8e7d8e7dc9d321e909f4ac81a91aaCAS |

Kitaji, H., Ookutsu, S., Sato, M., and Miyoshi, K. (2015). A new rolling culture-based in vitro fertilization system capable of reducing polyspermy in porcine oocytes. Anim. Sci. J. 86, 494–498.
A new rolling culture-based in vitro fertilization system capable of reducing polyspermy in porcine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXntFGlt7k%3D&md5=6f2d3f4ff6c2b960c99dac2e1822e4dfCAS |

Li, Y., Day, M. L., and O’Neill, C. (2007). Autocrine activation of ion currents in the two-cell mouse embryo. Exp. Cell Res. 313, 2786–2794.
Autocrine activation of ion currents in the two-cell mouse embryo.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXosFyqsLg%3D&md5=46de8219882d79f470b724a00d4d2b87CAS |

Li, R., Liu, Y., Pedersen, H. S., Kragh, P. M., and Callesen, H. (2013). Development and quality of porcine parthenogenetically activated embryos after removal of zona pellucida. Theriogenology 80, 58–64.
Development and quality of porcine parthenogenetically activated embryos after removal of zona pellucida.Crossref | GoogleScholarGoogle Scholar |

Li, J., Pedersen, H. S., Li, R., Adamsen, J., Liu, Y., Schmidt, M., Purup, S., and Callesen, H. (2014a). Developmental potential of pig embryos reconstructed by use of sow versus pre-pubertal gilt oocytes after somatic cell nuclear transfer. Zygote 22, 356–365.
Developmental potential of pig embryos reconstructed by use of sow versus pre-pubertal gilt oocytes after somatic cell nuclear transfer.Crossref | GoogleScholarGoogle Scholar |

Li, R., Pedersen, K. S., Liu, Y., Pedersen, H. S., Laegdsmand, M., Rickelt, L. F., Kuhl, M., and Callesen, H. (2014b). Effect of red light on the development and quality of mammalian embryos. J. Assist. Reprod. Genet. 31, 795–801.
Effect of red light on the development and quality of mammalian embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXhsFCqtb7I&md5=c4fcf5cb71614a041db601e463e1d428CAS |

Lin, L., Kragh, P. M., Purup, S., Kuwayama, M., Du, Y., Zhang, X., Yang, H., Bolund, L., Callesen, H., and Vajta, G. (2009). Osmotic stress induced by sodium chloride, sucrose or trehalose improves cryotolerance and developmental competence of porcine oocytes. Reprod. Fertil. Dev. 21, 338–344.
Osmotic stress induced by sodium chloride, sucrose or trehalose improves cryotolerance and developmental competence of porcine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtVensb8%3D&md5=d8c8572077b68af211fddb481e5a2e27CAS |

Liu, C., Wu, G. Q., Fu, X. W., Mo, X. H., Zhao, L. H., Hu, H. M., Zhu, S. E., and Hou, Y. P. (2015). The extracellular calcium-sensing receptor (CASR) regulates gonadotropins-induced meiotic maturation of porcine oocytes. Biol. Reprod. 93, 131.
The extracellular calcium-sensing receptor (CASR) regulates gonadotropins-induced meiotic maturation of porcine oocytes.Crossref | GoogleScholarGoogle Scholar |

Logan, J. E., and Roudebush, W. E. (2000). Platelet-activating factor increases intracellular calcium levels in preimplantation stage embryos. Early Pregnancy 4, 30–38.
| 1:STN:280:DC%2BD3Mnotl2itw%3D%3D&md5=9ed90b982620609e1e0cc9f532f9c2b0CAS |

Macías-García, B., Rocha, A., and González-Fernández, L. (2016). Extracellular calcium regulates protein tyrosine phosphorylation through calcium-sensing receptor (CaSR) in stallion sperm. Mol. Reprod. Dev. 83, 236–245.
Extracellular calcium regulates protein tyrosine phosphorylation through calcium-sensing receptor (CaSR) in stallion sperm.Crossref | GoogleScholarGoogle Scholar |

Marquez, B., and Suarez, S. S. (2008). Soluble adenylyl cyclase is required for activation of sperm but does not have a direct effect on hyperactivation. Reprod. Fertil. Dev. 20, 247–252.
Soluble adenylyl cyclase is required for activation of sperm but does not have a direct effect on hyperactivation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXislyqur0%3D&md5=05b3bd2dde6f178f4e33a37df2dbbf19CAS |

Martino, N. A., Lange-Consiglio, A., Cremonesi, F., Valentini, L., Caira, M., Guaricci, A. C., Ambruosi, B., Sciorsci, R. L., Lacalandra, G. M., Reshkin, S. J., and Dell’Aquila, M. E. (2011). Functional expression of the extracellular calcium sensing receptor (CaSR) in equine umbilical cord matrix size-sieved stem cells. PLoS One 6, e17714.
Functional expression of the extracellular calcium sensing receptor (CaSR) in equine umbilical cord matrix size-sieved stem cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXktVWhsb8%3D&md5=e0921dd69675b8df86997a226a58df5dCAS |

Mendoza, F. J., Perez-Marin, C. C., Garcia-Marin, L., Madueno, J. A., Henley, C., Aguilera-Tejero, E., and Rodriguez, M. (2012). Localization, distribution, and function of the calcium-sensing receptor in sperm. J. Androl. 33, 96–104.
Localization, distribution, and function of the calcium-sensing receptor in sperm.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhvVCntr8%3D&md5=6dc7eb2f31c871db61a0dde05a76b7bbCAS |

Michaut, M., Tomes, C. N., De Blas, G., Yunes, R., and Mayorga, L. S. (2000). Calcium-triggered acrosomal exocytosis in human spermatozoa requires the coordinated activation of Rab3A and N-ethylmaleimide-sensitive factor. Proc. Natl. Acad. Sci. USA 97, 9996–10001.
Calcium-triggered acrosomal exocytosis in human spermatozoa requires the coordinated activation of Rab3A and N-ethylmaleimide-sensitive factor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXmtlehs7k%3D&md5=3d51c921d5d0f8f05c73fe61f8afe077CAS |

Mizobuchi, M., Ogata, H., Hatamura, I., Saji, F., Koiwa, F., Kinugasa, E., Koshikawa, S., and Akizawa, T. (2007). Activation of calcium-sensing receptor accelerates apoptosis in hyperplastic parathyroid cells. Biochem. Biophys. Res. Commun. 362, 11–16.
Activation of calcium-sensing receptor accelerates apoptosis in hyperplastic parathyroid cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXpsFKlurw%3D&md5=f100276da29b30b2df56706135c34ef3CAS |

Nakamura, A., Hotsubo, T., Kobayashi, K., Mochizuki, H., Ishizu, K., and Tajima, T. (2013). Loss-of-function and gain-of-function mutations of calcium-sensing receptor: functional analysis and the effect of allosteric modulators NPS R-568 and NPS 2143. J. Clin. Endocrinol. Metab. 98, E1692–E1701.
| 1:CAS:528:DC%2BC3sXhs1elsLbK&md5=68fc62ecf446210d3eaca541c715027cCAS |

Pakrasi, P. L., and Dey, S. K. (1984). Role of calmodulin in blastocyst formation in the mouse. J. Reprod. Fertil. 71, 513–517.
Role of calmodulin in blastocyst formation in the mouse.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL2cXlt1Orur8%3D&md5=43047ba7898137085db4e3043598f0edCAS |

Pedersen, H. S., Liu, Y., Li, R., Purup, S., Lovendahl, P., Holm, P., Hyttel, P., and Callesen, H. (2015). Selection of pre- versus postpubertal pig oocytes for parthenogenetic activation and somatic cell nuclear transfer. Reprod. Fertil. Dev. 27, 544–550.
Selection of pre- versus postpubertal pig oocytes for parthenogenetic activation and somatic cell nuclear transfer.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXksVSltrs%3D&md5=3a68870382bca7e4ce3a76c9b21615eaCAS |

Rahman, M. S., Kwon, W. S., and Pang, M. G. (2014). Calcium influx and male fertility in the context of the sperm proteome: an update. BioMed Res. Int. 2014, 841615.
Calcium influx and male fertility in the context of the sperm proteome: an update.Crossref | GoogleScholarGoogle Scholar |

Ray, K. (2015). Calcium-sensing receptor: trafficking, endocytosis, recycling, and importance of interacting proteins. Prog. Mol. Biol. Transl. Sci. 132, 127–150.
Calcium-sensing receptor: trafficking, endocytosis, recycling, and importance of interacting proteins.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC28XhsFKmsbnO&md5=0874b66196631d8dfbe35d1a4028f268CAS |

Riley, J. K., Carayannopoulos, M. O., Wyman, A. H., Chi, M., Ratajczak, C. K., and Moley, K. H. (2005). The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo. Dev. Biol. 284, 377–386.
The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXpt1eksrY%3D&md5=c6da804de4996e0964fbf3f68d292285CAS |

Schuffner, A. A., Bastiaan, H. S., Duran, H. E., Lin, Z. Y., Morshedi, M., Franken, D. R., and Oehninger, S. (2002). Zona pellucida-induced acrosome reaction in human sperm: dependency on activation of pertussis toxin-sensitive G(i) protein and extracellular calcium, and priming effect of progesterone and follicular fluid. Mol. Hum. Reprod. 8, 722–727.
Zona pellucida-induced acrosome reaction in human sperm: dependency on activation of pertussis toxin-sensitive G(i) protein and extracellular calcium, and priming effect of progesterone and follicular fluid.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XmvFOrsb0%3D&md5=6249c3faa8587b1b0e5d7fbf85467f5cCAS |

Secher, J. O., Freude, K. K., Li, R., and Callesen, H. (2015). Optimization of three-dimensional imaging on in vitro produced porcine blastocysts and chimeras for stem cell testing: a technology report. Stem Cells Dev. 24, 1141–1145.
Optimization of three-dimensional imaging on in vitro produced porcine blastocysts and chimeras for stem cell testing: a technology report.Crossref | GoogleScholarGoogle Scholar |

Singh, R., Kumar, K., Mahapatra, P. S., Kumar, M., Agarwal, P., Bhure, S. K., Malakar, D., Bhanja, S. K., and Bag, S. (2014). Microarray analysis of gene expression in parthenotes and in vitro-derived goat embryos. Theriogenology 81, 854–860.
Microarray analysis of gene expression in parthenotes and in vitro-derived goat embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXitV2isrw%3D&md5=444f93ea4dde7f91a36dd8bed0f08365CAS |

Sozen, B., Ozturk, S., Yaba, A., and Demir, N. (2015). The p38 MAPK signalling pathway is required for glucose metabolism, lineage specification and embryo survival during mouse preimplantation development. Mech. Dev. 138, 375–398.
The p38 MAPK signalling pathway is required for glucose metabolism, lineage specification and embryo survival during mouse preimplantation development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXpsVWntrg%3D&md5=f23ade79785cd6eea6560b91dfabcf7bCAS |

Sun, Q. Y. (2003). Cellular and molecular mechanisms leading to cortical reaction and polyspermy block in mammalian eggs. Microsc. Res. Tech. 61, 342–348.
Cellular and molecular mechanisms leading to cortical reaction and polyspermy block in mammalian eggs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXls1Gnu7k%3D&md5=3af58b562ea1787f0c0bc5345de994f8CAS |

Suzuki, H., Saito, Y., Kagawa, N., and Yang, X. (2003). In vitro fertilization and polyspermy in the pig: factors affecting fertilization rates and cytoskeletal reorganization of the oocyte. Microsc. Res. Tech. 61, 327–334.
In vitro fertilization and polyspermy in the pig: factors affecting fertilization rates and cytoskeletal reorganization of the oocyte.Crossref | GoogleScholarGoogle Scholar |

Tfelt-Hansen, J., Chattopadhyay, N., Yano, S., Kanuparthi, D., Rooney, P., Schwarz, P., and Brown, E. M. (2004). Calcium-sensing receptor induces proliferation through p38 mitogen-activated protein kinase and phosphatidylinositol 3-kinase but not extracellularly regulated kinase in a model of humoral hypercalcemia of malignancy. Endocrinology 145, 1211–1217.
Calcium-sensing receptor induces proliferation through p38 mitogen-activated protein kinase and phosphatidylinositol 3-kinase but not extracellularly regulated kinase in a model of humoral hypercalcemia of malignancy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXhs1Wisbo%3D&md5=c0264ab4e882e00366be7074dc4cfc31CAS |

Thomsen, A. R., Hvidtfeldt, M., and Brauner-Osborne, H. (2012). Biased agonism of the calcium-sensing receptor. Cell Calcium 51, 107–116.
Biased agonism of the calcium-sensing receptor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xit12jt7g%3D&md5=0c3dac52405b542da7b6b5c95f27b4efCAS |

Visconti, P. E., Westbrook, V. A., Chertihin, O., Demarco, I., Sleight, S., and Diekman, A. B. (2002). Novel signaling pathways involved in sperm acquisition of fertilizing capacity. J. Reprod. Immunol. 53, 133–150.
Novel signaling pathways involved in sperm acquisition of fertilizing capacity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXos1Kjtrw%3D&md5=658694f7b08f56ba3b3833aad9ee34bcCAS |

Wu, Q. Y., Sun, M. R., Wu, C. L., Li, Y., Du, J. J., Zeng, J. Y., Bi, H. L., and Sun, Y. H. (2015). Activation of calcium-sensing receptor increases TRPC3/6 expression in T lymphocyte in sepsis. Mol. Immunol. 64, 18–25.
Activation of calcium-sensing receptor increases TRPC3/6 expression in T lymphocyte in sepsis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhvVKlt7zN&md5=4c939f65935dd1342594697d9fce7cf2CAS |

Xia, J., Reigada, D., Mitchell, C. H., and Ren, D. (2007). CATSPER channel-mediated Ca2+ entry into mouse sperm triggers a tail-to-head propagation. Biol. Reprod. 77, 551–559.
CATSPER channel-mediated Ca2+ entry into mouse sperm triggers a tail-to-head propagation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXpvFCrurk%3D&md5=27ea24d0ca487b9df7519568a7d2c0f8CAS |

Yoshioka, K., Suzuki, C., Tanaka, A., Anas, I. M., and Iwamura, S. (2002). Birth of piglets derived from porcine zygotes cultured in a chemically defined medium. Biol. Reprod. 66, 112–119.
Birth of piglets derived from porcine zygotes cultured in a chemically defined medium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xht1yksQ%3D%3D&md5=9a6c69dd84f74136f8a315e08dcb5d0bCAS |

Zhen, Y., Ding, C., Sun, J., Wang, Y., Li, S., and Dong, L. (2016). Activation of the calcium-sensing receptor promotes apoptosis by modulating the JNK/p38 MAPK pathway in focal cerebral ischemia-reperfusion in mice. Am. J. Transl. Res. 8, 911–921.