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

Role of reproductive fluids and extracellular vesicles in embryo–maternal interaction during early pregnancy in cattle

Yulia N. Cajas A B , Karina Cañón-Beltrán A C , María Gemma Millán de la Blanca A , José M. Sánchez A , Beatriz Fernandez-Fuertes A , Encina M. González D and Dimitrios Rizos https://orcid.org/0000-0001-6813-3940 A *
+ Author Affiliations
- Author Affiliations

A Department of Animal Reproduction, National Center Institute for Agriculture and Food Research and Technology (CSIC-INIA), Ctra de la Coruña KM 5.9, 28040 Madrid, Spain.

B Laboratorio de Biotecnología de la Reproducción Animal, Facultad de Ciencias Agropecuarias, Universidad de Cuenca (UC), EC010205 Cuenca, Ecuador.

C Facultad de Ciencias Agrarias y Ambientales, Programa de Medicina Veterinaria, Fundación Universitaria Juan de Castellanos (JdC), 150001 Tunja, Colombia.

D Department of Anatomy and Embryology, Veterinary Faculty, Complutense University of Madrid (UCM), 28040 Madrid, Spain.

* Correspondence to: drizos@inia.es

Reproduction, Fertility and Development 34(2) 117-138 https://doi.org/10.1071/RD21275
Published online: 2 November 2021

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing on behalf of the IETS

Abstract

The coordinated interaction between the developing embryo and the maternal reproductive tract is essential for the establishment and maintenance of pregnancy in mammals. An early cross-talk is established between the oviduct/uterus and the gametes and embryo. This dialogue will shape the microenvironment in which gamete transport, fertilisation, and early embryonic development occur. Due to the small size of the gametes and the early embryo relative to the volume of the oviductal and uterine lumina, collection of tissue and fluid adjacent to these cells is challenging in cattle. Thus, the combination of in vivo and in vitro models seems to be the most appropriate approach to better understand this fine dialogue. In this respect, the aim of this review is to summarise the recent findings in relation to gamete/embryo–maternal interaction during the pre-elongation period.

Keywords: cattle, EV biomarkers, extracellular vesicles, miRNA, mRNA, oviductal fluid, oviductosomes, uterine fluid, uterosomes.


References

Aguilar, J, and Reyley, M (2005). The uterine tubal fluid: secretion, composition and biological effects. Animal Reproduction 2, 91–105.

Alcântara-Neto, AS, Fernandez-Rufete, M, Corbin, E, Tsikis, G, Uzbekov, R, Garanina, AS, Coy, P, Almiñana, C, and Mermillod, P (2020a). Oviduct fluid extracellular vesicles regulate polyspermy during porcine in vitro fertilisation. Reproduction, Fertility and Development 32, 409–418.
Oviduct fluid extracellular vesicles regulate polyspermy during porcine in vitro fertilisation.Crossref | GoogleScholarGoogle Scholar |

Alcântara-Neto, AS, Schmaltz, L, Caldas, E, Blache, M-C, Mermillod, P, and Almiñana, C (2020b). Porcine oviductal extracellular vesicles interact with gametes and regulate sperm motility and survival. Theriogenology 155, 240–255.
Porcine oviductal extracellular vesicles interact with gametes and regulate sperm motility and survival.Crossref | GoogleScholarGoogle Scholar | 32791377PubMed |

Alexandre, H (2001). A history of mammalian embryological research. The International Journal of Developmental Biology 45, 457–467.
| 11417885PubMed |

Algarra, B, Maillo, V, Avilés, M, Gutiérrez-Adán, A, Rizos, D, and Jiménez-Movilla, M (2018). Effects of recombinant OVGP1 protein on in vitro bovine embryo development. Journal of Reproduction and Development 64, 433–443.
Effects of recombinant OVGP1 protein on in vitro bovine embryo development.Crossref | GoogleScholarGoogle Scholar |

Almiñana, C, and Bauersachs, S (2019). Extracellular vesicles in the oviduct: progress, challenges and implications for the reproductive success. Bioengineering 6, 32.
Extracellular vesicles in the oviduct: progress, challenges and implications for the reproductive success.Crossref | GoogleScholarGoogle Scholar |

Almiñana, C, Corbin, E, Tsikis, G, Alcântara-Neto, AS, Labas, V, Reynaud, K, Galio, L, Uzbekov, R, Garanina, AS, Druart, X, and Mermillod, P (2017). Oviduct extracellular vesicles protein content and their role during oviduct–embryo cross-talk. Reproduction 154, 253–268.
Oviduct extracellular vesicles protein content and their role during oviduct–embryo cross-talk.Crossref | GoogleScholarGoogle Scholar |

Almiñana, C, Heath, PR, Wilkinson, S, Sanchez-Osorio, J, Cuello, C, Parrilla, I, Gil, MA, Vazquez, JL, Vazquez, JM, Roca, J, Martinez, EA, and Fazeli, A (2012). Early developing pig embryos mediate their own environment in the maternal tract. PLoS ONE 7, e33625.
Early developing pig embryos mediate their own environment in the maternal tract.Crossref | GoogleScholarGoogle Scholar | 22470458PubMed |

Almiñana, C, Tsikis, G, Labas, V, Uzbekov, R, da Silveira, JC, Bauersachs, S, and Mermillod, P (2018). Deciphering the oviductal extracellular vesicles content across the estrous cycle: implications for the gametes-oviduct interactions and the environment of the potential embryo. BMC Genomics 19, 622.
Deciphering the oviductal extracellular vesicles content across the estrous cycle: implications for the gametes-oviduct interactions and the environment of the potential embryo.Crossref | GoogleScholarGoogle Scholar | 30134841PubMed |

Araújo, ER, Sponchiado, M, Pugliesi, G, Van Hoeck, V, Mesquita, FS, Membrive, CMB, and Binelli, M (2014). Spatio-specific regulation of endocrine-responsive gene transcription by periovulatory endocrine profiles in the bovine reproductive tract. Reproduction, Fertility and Development 28, 1533–1544.
Spatio-specific regulation of endocrine-responsive gene transcription by periovulatory endocrine profiles in the bovine reproductive tract.Crossref | GoogleScholarGoogle Scholar |

Ardon, F, Markello, RD, Hu, L, Deutsch, ZI, Tung, C-K, Wu, M, and Suarez, SS (2016). Dynamics of bovine sperm interaction with epithelium differ between oviductal isthmus and ampulla. Biology of Reproduction 95, 90.
Dynamics of bovine sperm interaction with epithelium differ between oviductal isthmus and ampulla.Crossref | GoogleScholarGoogle Scholar | 27605344PubMed |

Asaadi, A, Dolatabad, NA, Atashi, H, Raes, A, Van Damme, P, Hoelker, M, Hendrix, A, Pascottini, OB, Van Soom, A, Kafi, M, and Pavani, KC (2021). Extracellular vesicles from follicular and ampullary fluid isolated by density gradient ultracentrifugation improve bovine embryo development and quality. International Journal of Molecular Sciences 22, 578.
Extracellular vesicles from follicular and ampullary fluid isolated by density gradient ultracentrifugation improve bovine embryo development and quality.Crossref | GoogleScholarGoogle Scholar |

Asaadi, A, Kafi, M, Atashi, H, Azari, M, and Hostens, M (2019). Frozen–thawed ampullary cell monolayer improves bovine embryo in vitro development and quality. Zygote 27, 337–346.
Frozen–thawed ampullary cell monolayer improves bovine embryo in vitro development and quality.Crossref | GoogleScholarGoogle Scholar | 31405390PubMed |

Bagés-Arnal, S, Sánchez, JM, Fernandez-Fuertes, B, McDonald, M, Behura, SK, Spencer, TE, Fair, T, and Lonergan, P (2020). Location relative to the corpus luteum affects bovine endometrial response to a conceptus. Reproduction 159, 643–657.
Location relative to the corpus luteum affects bovine endometrial response to a conceptus.Crossref | GoogleScholarGoogle Scholar | 32168470PubMed |

Banliat, C, Le Bourhis, D, Bernardi, O, Tomas, D, Labas, V, Salvetti, P, Guyonnet, B, Mermillod, P, and Saint-Dizier, M (2020). Oviduct fluid extracellular vesicles change the phospholipid composition of bovine embryos developed in vitro. International Journal of Molecular Sciences 21, 5326.
Oviduct fluid extracellular vesicles change the phospholipid composition of bovine embryos developed in vitro.Crossref | GoogleScholarGoogle Scholar |

Barrera, AD, García, EV, Hamdi, M, Sánchez-Calabuig, MJ, López-Cardona, ÁP, Balvís, NF, Rizos, D, and Gutiérrez-Adán, A (2017). Embryo culture in presence of oviductal fluid induces DNA methylation changes in bovine blastocysts. Reproduction 154, 1–12.
Embryo culture in presence of oviductal fluid induces DNA methylation changes in bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 28408706PubMed |

Bauersachs, S, Blum, H, Mallok, S, Wenigerkind, H, Rief, S, Prelle, K, and Wolf, E (2003). Regulation of ipsilateral and contralateral bovine oviduct epithelial cell function in the postovulation period: a transcriptomics approach. Biology of Reproduction 68, 1170–1177.
Regulation of ipsilateral and contralateral bovine oviduct epithelial cell function in the postovulation period: a transcriptomics approach.Crossref | GoogleScholarGoogle Scholar | 12606461PubMed |

Bauersachs, S, Mermillod, P, and Almiñana, C (2020). The oviductal extracellular vesicles’ RNA cargo regulates the bovine embryonic transcriptome. International Journal of Molecular Sciences 21, 1303.
The oviductal extracellular vesicles’ RNA cargo regulates the bovine embryonic transcriptome.Crossref | GoogleScholarGoogle Scholar |

Bauersachs, S, Rehfeld, S, Ulbrich, SE, Mallok, S, Prelle, K, Wenigerkind, H, Einspanier, R, Blum, H, and Wolf, E (2004). Monitoring gene expression changes in bovine oviduct epithelial cells during the oestrous cycle. Journal of Molecular Endocrinology 32, 449–466.
Monitoring gene expression changes in bovine oviduct epithelial cells during the oestrous cycle.Crossref | GoogleScholarGoogle Scholar | 15072551PubMed |

Bauersachs, S, Ulbrich, SE, Reichenbach, H-D, Reichenbach, M, Büttner, M, Meyer, HHD, Spencer, TE, Minten, M, Sax, G, Winter, G, and Wolf, E (2012). Comparison of the effects of early pregnancy with human interferon, alpha 2 (IFNA2), on gene expression in bovine endometrium. Biology of Reproduction 86, 46.
Comparison of the effects of early pregnancy with human interferon, alpha 2 (IFNA2), on gene expression in bovine endometrium.Crossref | GoogleScholarGoogle Scholar | 22034527PubMed |

Bazer, FW, Song, G, Kim, J, Erikson, DW, Johnson, GA, Burghardt, RC, Gao, H, Carey Satterfield, M, Spencer, TE, and Wu, G (2012). Mechanistic mammalian target of rapamycin (MTOR) cell signaling: Effects of select nutrients and secreted phosphoprotein 1 on development of mammalian conceptuses. Molecular and Cellular Endocrinology 354, 22–33.
Mechanistic mammalian target of rapamycin (MTOR) cell signaling: Effects of select nutrients and secreted phosphoprotein 1 on development of mammalian conceptuses.Crossref | GoogleScholarGoogle Scholar | 21907263PubMed |

Bhusane, K, Bhutada, S, Chaudhari, U, Savardekar, L, Katkam, R, and Sachdeva, G (2016). Secrets of endometrial receptivity: some are hidden in uterine secretome. American Journal of Reproductive Immunology 75, 226–236.
Secrets of endometrial receptivity: some are hidden in uterine secretome.Crossref | GoogleScholarGoogle Scholar | 26865379PubMed |

Binder, NK, Evans, J, Gardner, DK, Salamonsen, LA, and Hannan, NJ (2014). Endometrial signals improve embryo outcome: functional role of vascular endothelial growth factor isoforms on embryo development and implantation in mice. Human Reproduction 29, 2278–2286.
Endometrial signals improve embryo outcome: functional role of vascular endothelial growth factor isoforms on embryo development and implantation in mice.Crossref | GoogleScholarGoogle Scholar | 25124669PubMed |

Binelli, M, Scolari, SC, Pugliesi, G, Van Hoeck, V, Gonella-Diaza, AM, Andrade, SCS, Gasparin, GR, and Coutinho, LL (2015). The transcriptome signature of the receptive bovine uterus determined at early gestation. PLoS ONE 10, e0122874.
The transcriptome signature of the receptive bovine uterus determined at early gestation.Crossref | GoogleScholarGoogle Scholar | 25849079PubMed |

Bridi, A, Andrade, GM, Del Collado, M, Sangalli, JR, de Ávila, ACFCM, Motta, IG, da Silva, JCB, Pugliesi, G, Silva, LA, Meirelles, FV, da Silveira, JC, and Perecin, F (2021). Small extracellular vesicles derived from in vivo- or in vitro-produced bovine blastocysts have different miRNAs profiles—implications for embryo–maternal recognition. Molecular Reproduction and Development 88, 628–643.
Small extracellular vesicles derived from in vivo- or in vitro-produced bovine blastocysts have different miRNAs profiles—implications for embryo–maternal recognition.Crossref | GoogleScholarGoogle Scholar | 34402123PubMed |

Bridi, A, Perecin, F, and da Silveira, JC (2020). Extracellular vesicles mediated early embryo–maternal interactions. International Journal of Molecular Sciences 21, 1163.
Extracellular vesicles mediated early embryo–maternal interactions.Crossref | GoogleScholarGoogle Scholar |

Brower, LK, and Anderson, E (1969). Cytological events associated with the secretory process in the rabbit oviduct. Biology of Reproduction 1, 130–148.
Cytological events associated with the secretory process in the rabbit oviduct.Crossref | GoogleScholarGoogle Scholar | 4257488PubMed |

Buhi, WC, Alvarez, IM, Sudhipong, V, and Dones-Smith, MM (1990). Identification and characterization of de novo-synthesized porcine oviductal secretory proteins. Biology of Reproduction 43, 929–938.
Identification and characterization of de novo-synthesized porcine oviductal secretory proteins.Crossref | GoogleScholarGoogle Scholar | 2291929PubMed |

Burns, G, Brooks, K, Wildung, M, Navakanitworakul, R, Christenson, LK, and Spencer, TE (2014). Extracellular vesicles in luminal fluid of the ovine uterus. PLoS ONE 9, e90913.
Extracellular vesicles in luminal fluid of the ovine uterus.Crossref | GoogleScholarGoogle Scholar | 24614226PubMed |

Burns, GW, Brooks, KE, O’Neil, EV, Hagen, DE, Behura, SK, and Spencer, TE (2018). Progesterone effects on extracellular vesicles in the sheep uterus. Biology of Reproduction 98, 612–622.
Progesterone effects on extracellular vesicles in the sheep uterus.Crossref | GoogleScholarGoogle Scholar | 29346527PubMed |

Burns, GW, Brooks, KE, and Spencer, TE (2016). Extracellular vesicles originate from the conceptus and uterus during early pregnancy in sheep. Biology of Reproduction 94, 56.
Extracellular vesicles originate from the conceptus and uterus during early pregnancy in sheep.Crossref | GoogleScholarGoogle Scholar | 26819476PubMed |

Cañón-Beltrán K, Hamdi M, Mazzarella R, Cajas YN, Leal CLV, Gutiérrez-Adán A, González EM, da Silveira JC, Rizos D (2021) Isolation, characterization, and microRNA analysis of extracellular vesicles from bovine oviduct and uterine fluids. In ‘Next generation culture platforms for reliable in vitro models’. Methods in molecular biology. (Eds TAL Brevini, A Fazeli, K Turksen) pp. 219–238. (Springer: New York, NY, USA)
| Crossref |

Capra, E, and Lange-Consiglio, A (2020). The biological function of extracellular vesicles during fertilization, early embryo–maternal crosstalk and their involvement in reproduction: review and overview. Biomolecules 10, 1510.
The biological function of extracellular vesicles during fertilization, early embryo–maternal crosstalk and their involvement in reproduction: review and overview.Crossref | GoogleScholarGoogle Scholar |

Cebrian-Serrano, A, Salvador, I, García-Roselló, E, Pericuesta, E, Pérez-Cerezales, S, Gutierrez-Adán, A, Coy, P, and Silvestre, MA (2013). Effect of the bovine oviductal fluid on in vitro fertilization, development and gene expression of in vitro-produced bovine blastocysts. Reproduction in Domestic Animals 48, 331–338.
Effect of the bovine oviductal fluid on in vitro fertilization, development and gene expression of in vitro-produced bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 22908847PubMed |

Cerny, KL, Garrett, E, Walton, AJ, Anderson, LH, and Bridges, PJ (2015). A transcriptomal analysis of bovine oviductal epithelial cells collected during the follicular phase versus the luteal phase of the estrous cycle. Reproductive Biology and Endocrinology 13, 84.
A transcriptomal analysis of bovine oviductal epithelial cells collected during the follicular phase versus the luteal phase of the estrous cycle.Crossref | GoogleScholarGoogle Scholar | 26242217PubMed |

Corcoran, D, Rizos, D, Fair, T, Evans, ACO, and Lonergan, P (2007). Temporal expression of transcripts related to embryo quality in bovine embryos cultured from the two-cell to blastocyst stage in vitro or in vivo. Molecular Reproduction and Development 74, 972–977.
Temporal expression of transcripts related to embryo quality in bovine embryos cultured from the two-cell to blastocyst stage in vitro or in vivo.Crossref | GoogleScholarGoogle Scholar | 17219429PubMed |

Cordova, A, Perreau, C, Uzbekova, S, Ponsart, C, Locatelli, Y, and Mermillod, P (2014). Development rate and gene expression of IVP bovine embryos cocultured with bovine oviduct epithelial cells at early or late stage of preimplantation development. Theriogenology 81, 1163–1173.
Development rate and gene expression of IVP bovine embryos cocultured with bovine oviduct epithelial cells at early or late stage of preimplantation development.Crossref | GoogleScholarGoogle Scholar | 24629595PubMed |

Coy, P, Cánovas, S, Mondéjar, I, Saavedra, MD, Romar, R, Grullón, L, Matás, C, and Avilés, M (2008). Oviduct-specific glycoprotein and heparin modulate sperm-zona pellucida interaction during fertilization and contribute to the control of polyspermy. Proceedings of the National Academy of Sciences of the United States of America 105, 15809–15814.
Oviduct-specific glycoprotein and heparin modulate sperm-zona pellucida interaction during fertilization and contribute to the control of polyspermy.Crossref | GoogleScholarGoogle Scholar | 18838686PubMed |

Coy, P, Jiménez-Movilla, M, García-Vázquez, FA, Mondéjar, I, Grullón, L, and Romar, R (2012). Oocytes use the plasminogen-plasmin system to remove supernumerary spermatozoa. Human Reproduction 27, 1985–1993.
Oocytes use the plasminogen-plasmin system to remove supernumerary spermatozoa.Crossref | GoogleScholarGoogle Scholar | 22556378PubMed |

Dadashpour Davachi, N, Kohram, H, Zare Shahneh, A, Zhandi, M, Goudarzi, A, Fallahi, R, Masoudi, R, Yousefi, AR, and Bartlewski, PM (2017). The effect of conspecific ampulla oviductal epithelial cells during in vitro maturation on oocyte developmental competence and maturation-promoting factor (MPF) activity in sheep. Theriogenology 88, 207–214.
The effect of conspecific ampulla oviductal epithelial cells during in vitro maturation on oocyte developmental competence and maturation-promoting factor (MPF) activity in sheep.Crossref | GoogleScholarGoogle Scholar | 28234231PubMed |

Dadashpour Davachi, N, Zare Shahneh, A, Kohram, H, Zhandi, M, Shamsi, H, Hajiyavand, AM, and Saadat, M (2016). Differential influence of ampullary and isthmic derived epithelial cells on zona pellucida hardening and in vitro fertilization in ovine. Reproductive Biology 16, 61–69.
Differential influence of ampullary and isthmic derived epithelial cells on zona pellucida hardening and in vitro fertilization in ovine.Crossref | GoogleScholarGoogle Scholar | 26952755PubMed |

Diskin, MG, Waters, SM, Parr, MH, and Kenny, DA (2016). Pregnancy losses in cattle: potential for improvement. Reproduction, Fertility and Development 28, 83–93.
Pregnancy losses in cattle: potential for improvement.Crossref | GoogleScholarGoogle Scholar |

Dissanayake, K, Nõmm, M, Lättekivi, F, Ord, J, Ressaissi, Y, Godakumara, K, Reshi, QU A, Viil, J, Jääger, K, Velthut-Meikas, A, Salumets, A, Jaakma, Ü, and Fazeli, A (2021). Oviduct as a sensor of embryo quality: deciphering the extracellular vesicle (EV)-mediated embryo–maternal dialogue. Journal of Molecular Medicine 99, 685–697.
Oviduct as a sensor of embryo quality: deciphering the extracellular vesicle (EV)-mediated embryo–maternal dialogue.Crossref | GoogleScholarGoogle Scholar | 33512581PubMed |

Dissanayake, K, Nõmm, M, Lättekivi, F, Ressaissi, Y, Godakumara, K, Lavrits, A, Midekessa, G, Viil, J, Bæk, R, Jørgensen, MM, Bhattacharjee, S, Andronowska, A, Salumets, A, Jaakma, Ü, and Fazeli, A (2020). Individually cultured bovine embryos produce extracellular vesicles that have the potential to be used as non-invasive embryo quality markers. Theriogenology 149, 104–116.
Individually cultured bovine embryos produce extracellular vesicles that have the potential to be used as non-invasive embryo quality markers.Crossref | GoogleScholarGoogle Scholar | 32259747PubMed |

Driver, AM, Peñagaricano, F, Huang, W, Ahmad, KR, Hackbart, KS, Wiltbank, MC, and Khatib, H (2012). RNA-Seq analysis uncovers transcriptomic variations between morphologically similar in vivo- and in vitro-derived bovine blastocysts. BMC Genomics 13, 118.
RNA-Seq analysis uncovers transcriptomic variations between morphologically similar in vivo- and in vitro-derived bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 22452724PubMed |

Ealy, AD, Wooldridge, LK, and McCoski, SR (2019). Board invited review: post-transfer consequences of in vitro-produced embryos in cattle. Journal of Animal Science 97, 2555–2568.
Board invited review: post-transfer consequences of in vitro-produced embryos in cattle.Crossref | GoogleScholarGoogle Scholar | 30968113PubMed |

Eisenbach, M (1999). Mammalian sperm chemotaxis and its association with capacitation. Developmental Genetics 25, 87–94.
Mammalian sperm chemotaxis and its association with capacitation.Crossref | GoogleScholarGoogle Scholar | 10440842PubMed |

Eisenbach, M, and Giojalas, LC (2006). Sperm guidance in mammals – an unpaved road to the egg. Nature Reviews Molecular Cell Biology 7, 276–285.
Sperm guidance in mammals – an unpaved road to the egg.Crossref | GoogleScholarGoogle Scholar | 16607290PubMed |

Fang, F, Li, Z, Yu, J, Long, Y, Zhao, Q, Ding, X, Wu, L, Shao, S, Zhang, L, and Xiang, W (2021). MicroRNAs secreted by human embryos could be potential biomarkers for clinical outcomes of assisted reproductive technology. Journal of Advanced Research 31, 25–34.
MicroRNAs secreted by human embryos could be potential biomarkers for clinical outcomes of assisted reproductive technology.Crossref | GoogleScholarGoogle Scholar | 34194830PubMed |

Faulkner, S, Elia, G, O’Boyle, P, Dunn, M, and Morris, D (2013). Composition of the bovine uterine proteome is associated with stage of cycle and concentration of systemic progesterone. Proteomics 13, 3333–3353.
Composition of the bovine uterine proteome is associated with stage of cycle and concentration of systemic progesterone.Crossref | GoogleScholarGoogle Scholar | 24115321PubMed |

Fereshteh, Z, Schmidt, SA, Al-Dossary, AA, Accerbi, M, Arighi, C, Cowart, J, Song, JL, Green, PJ, Choi, K, Yoo, S, and Martin-DeLeon, PA (2018). Murine oviductosomes (OVS) microRNA profiling during the estrous cycle: delivery of OVS-borne microRNAs to sperm where miR-34c-5p localizes at the centrosome. Scientific Reports 8, 16094.
Murine oviductosomes (OVS) microRNA profiling during the estrous cycle: delivery of OVS-borne microRNAs to sperm where miR-34c-5p localizes at the centrosome.Crossref | GoogleScholarGoogle Scholar | 30382141PubMed |

Ferraz, M, Nagashima, JB, Noonan, MJ, Crosier, AE, and Songsasen, N (2020). Oviductal extracellular vesicles improve post-thaw sperm function in red wolves and cheetahs. International Journal of Molecular Sciences 21, 3733.
Oviductal extracellular vesicles improve post-thaw sperm function in red wolves and cheetahs.Crossref | GoogleScholarGoogle Scholar |

Ferraz, MdAMM, Carothers, A, Dahal, R, Noonan, MJ, and Songsasen, N (2019). Oviductal extracellular vesicles interact with the spermatozoon’s head and mid-piece and improves its motility and fertilizing ability in the domestic cat. Scientific Reports 9, 9484.
Oviductal extracellular vesicles interact with the spermatozoon’s head and mid-piece and improves its motility and fertilizing ability in the domestic cat.Crossref | GoogleScholarGoogle Scholar |

Flesch, FM, Brouwers, JF, Nievelstein, PF, Verkleij, AJ, van Golde, LM, Colenbrander, B, and Gadella, BM (2001). Bicarbonate stimulated phospholipid scrambling induces cholesterol redistribution and enables cholesterol depletion in the sperm plasma membrane. Journal of Cell Science 114, 3543–3555.
Bicarbonate stimulated phospholipid scrambling induces cholesterol redistribution and enables cholesterol depletion in the sperm plasma membrane.Crossref | GoogleScholarGoogle Scholar | 11682613PubMed |

Fontes, PK, Ereno, RL, Peixoto, AR, Carvalho, RF, Scarano, WR, Trinca, LA, Barros, CM, and Castilho, ACdS (2018). Can the antral follicular count modulate the gene expression of bovine oviducts in Aberdeen Angus and Nelore heifers? PLoS ONE 13, e0202017.
Can the antral follicular count modulate the gene expression of bovine oviducts in Aberdeen Angus and Nelore heifers?Crossref | GoogleScholarGoogle Scholar | 30157205PubMed |

Forde, N, Carter, F, Fair, T, Crowe, MA, Evans, ACO, Spencer, TE, Bazer, FW, McBride, R, Boland, MP, O’Gaora, P, Lonergan, P, and Roche, JF (2009). Progesterone-regulated changes in endometrial gene expression contribute to advanced conceptus development in cattle. Biology of Reproduction 81, 784–794.
Progesterone-regulated changes in endometrial gene expression contribute to advanced conceptus development in cattle.Crossref | GoogleScholarGoogle Scholar | 19553605PubMed |

Forde, N, Carter, F, Spencer, TE, Bazer, FW, Sandra, O, Mansouri-Attia, N, Okumu, LA, McGettigan, PA, Mehta, JP, McBride, R, O’Gaora, P, Roche, JF, and Lonergan, P (2011). Conceptus-induced changes in the endometrial transcriptome: how soon does the cow know she is pregnant? Biology of Reproduction 85, 144–156.
Conceptus-induced changes in the endometrial transcriptome: how soon does the cow know she is pregnant?Crossref | GoogleScholarGoogle Scholar | 21349821PubMed |

Franchi, A, Moreno-Irusta, A, Domínguez, EM, Adre, AJ, and Giojalas, LC (2020). Extracellular vesicles from oviductal isthmus and ampulla stimulate the induced acrosome reaction and signaling events associated with capacitation in bovine spermatozoa. Journal of Cellular Biochemistry 121, 2877–2888.
Extracellular vesicles from oviductal isthmus and ampulla stimulate the induced acrosome reaction and signaling events associated with capacitation in bovine spermatozoa.Crossref | GoogleScholarGoogle Scholar | 31692037PubMed |

Gad, A, Hoelker, M, Besenfelder, U, Havlicek, V, Cinar, U, Rings, F, Held, E, Dufort, I, Sirard, M-A, Schellander, K, and Tesfaye, D (2012a). Molecular mechanisms and pathways involved in bovine embryonic genome activation and their regulation by alternative in vivo and in vitro culture conditions. Biology of Reproduction 87, 100.
Molecular mechanisms and pathways involved in bovine embryonic genome activation and their regulation by alternative in vivo and in vitro culture conditions.Crossref | GoogleScholarGoogle Scholar | 22811576PubMed |

Gad, A, Schellander, K, Hoelker, M, and Tesfaye, D (2012b). Transcriptome profile of early mammalian embryos in response to culture environment. Animal Reproduction Science 134, 76–83.
Transcriptome profile of early mammalian embryos in response to culture environment.Crossref | GoogleScholarGoogle Scholar | 22917875PubMed |

Gallicano, GI (2001). Composition, regulation, and function of the cytoskeleton in mammalian eggs and embryos. Frontiers in Bioscience 6, D1089–D1108.
Composition, regulation, and function of the cytoskeleton in mammalian eggs and embryos.Crossref | GoogleScholarGoogle Scholar | 11532603PubMed |

García, EV, Hamdi, M, Barrera, AD, Sánchez-Calabuig, MJ, Gutiérrez-Adán, A, and Rizos, D (2017). Bovine embryo-oviduct interaction in vitro reveals an early cross talk mediated by BMP signaling. Reproduction 153, 631–643.
Bovine embryo-oviduct interaction in vitro reveals an early cross talk mediated by BMP signaling.Crossref | GoogleScholarGoogle Scholar | 28250237PubMed |

García, EV, Miceli, DC, Rizo, G, Valdecantos, PA, and Barrera, AD (2015). Effect of early addition of bone morphogenetic protein 5 (BMP5) to embryo culture medium on in vitro development and expression of developmentally important genes in bovine preimplantation embryos. Theriogenology 84, 589–599.
Effect of early addition of bone morphogenetic protein 5 (BMP5) to embryo culture medium on in vitro development and expression of developmentally important genes in bovine preimplantation embryos.Crossref | GoogleScholarGoogle Scholar | 26014926PubMed |

Gatien, J, Mermillod, P, Tsikis, G, Bernardi, O, Janati Idrissi, S, Uzbekov, R, Le Bourhis, D, Salvetti, P, Almiñana, C, and Saint-Dizier, M (2019). Metabolomic profile of oviductal extracellular vesicles across the estrous cycle in cattle. International Journal of Molecular Sciences 20, 6339.
Metabolomic profile of oviductal extracellular vesicles across the estrous cycle in cattle.Crossref | GoogleScholarGoogle Scholar |

Gellersen, B, Fernandes, MS, and Brosens, JJ (2009). Non-genomic progesterone actions in female reproduction. Human Reproduction Update 15, 119–138.
Non-genomic progesterone actions in female reproduction.Crossref | GoogleScholarGoogle Scholar | 18936037PubMed |

Ghersevich, S, Massa, E, and Zumoffen, C (2015). Oviductal secretion and gamete interaction. Reproduction 149, R1–R14.
Oviductal secretion and gamete interaction.Crossref | GoogleScholarGoogle Scholar | 25190504PubMed |

Giacomini, E, Vago, R, Sanchez, AM, Podini, P, Zarovni, N, Murdica, V, Rizzo, R, Bortolotti, D, Candiani, M, and Viganò, P (2017). Secretome of in vitro cultured human embryos contains extracellular vesicles that are uptaken by the maternal side. Scientific Reports 7, 5210.
Secretome of in vitro cultured human embryos contains extracellular vesicles that are uptaken by the maternal side.Crossref | GoogleScholarGoogle Scholar | 28701751PubMed |

Gómez, E, Correia-Álvarez, E, Caamaño, JN, Díez, C, Carrocera, S, Peynot, N, Martín, D, Giraud-Delville, C, Duranthon, V, Sandra, O, and Muñoz, M (2014). Hepatoma-derived growth factor: from the bovine uterus to the in vitro embryo culture. Reproduction 148, 353–365.
Hepatoma-derived growth factor: from the bovine uterus to the in vitro embryo culture.Crossref | GoogleScholarGoogle Scholar | 25009202PubMed |

Gonella-Diaza, AM, Andrade, SCdS, Sponchiado, M, Pugliesi, G, Mesquita, FS, Van Hoeck, V, Strefezzi, RdF, Gasparin, GR, Coutinho, LL, and Binelli, M (2015). Size of the ovulatory follicle dictates spatial differences in the oviductal transcriptome in Cattle. PloS ONE 10, e0145321.
Size of the ovulatory follicle dictates spatial differences in the oviductal transcriptome in Cattle.Crossref | GoogleScholarGoogle Scholar | 26699362PubMed |

Gonella-Diaza, AM, Lopes, E, Ribeiro da Silva, K, Perecin Nociti, R, Mamede Andrade, G, Atuesta-Bustos, JE, Coelho da Silveira, J, Vieira Meirelles, F, and Binelli, M (2021). Steroidal regulation of oviductal microRNAs is associated with microRNA-processing in beef cows. International Journal of Molecular Sciences 22, 953.
Steroidal regulation of oviductal microRNAs is associated with microRNA-processing in beef cows.Crossref | GoogleScholarGoogle Scholar | 33477993PubMed |

González-Brusi, L, Algarra, B, Moros-Nicolás, C, Izquierdo-Rico, MJ, Avilés, M, and Jiménez-Movilla, M (2020). A comparative view on the oviductal environment during the periconception period. Biomolecules 10, 1690.
A comparative view on the oviductal environment during the periconception period.Crossref | GoogleScholarGoogle Scholar |

Gray, CA, Taylor, KM, Ramsey, WS, Hill, JR, Bazer, FW, Bartol, FF, and Spencer, TE (2001). Endometrial glands are required for preimplantation conceptus elongation and survival. Biology of Reproduction 64, 1608–1613.
Endometrial glands are required for preimplantation conceptus elongation and survival.Crossref | GoogleScholarGoogle Scholar | 11369585PubMed |

Hamdi, M, Lopera-Vasquez, R, Maillo, V, Sanchez-Calabuig, MJ, Núnez, C, Gutierrez-Adan, A, and Rizos, D (2018). Bovine oviductal and uterine fluid support in vitro embryo development. Reproduction, Fertility and Development 30, 935–945.
Bovine oviductal and uterine fluid support in vitro embryo development.Crossref | GoogleScholarGoogle Scholar |

Hamdi, M, Mazzarella, R, Cañon-Beltrán, K, Cajas, YN, Leal, CLV, Gutiérrez-Adán, A, González, ME, da Silveira, JC, and Rizos, D (2021). 36 Analysis of miRNA content of oviduct and uterine extracellular vesicles across the bovine estrous cycle. Reproduction, Fertility and Development 33, 125–125.
36 Analysis of miRNA content of oviduct and uterine extracellular vesicles across the bovine estrous cycle.Crossref | GoogleScholarGoogle Scholar |

Hamdi, M, Sánchez-Calabuig, MJ, Rodríguez-Alonso, B, Arnal, SB, Roussi, K, Sturmey, R, Gutiérrez-Adán, A, Lonergan, P, and Rizos, D (2019). Gene expression and metabolic response of bovine oviduct epithelial cells to the early embryo. Reproduction 158, 85–94.
Gene expression and metabolic response of bovine oviduct epithelial cells to the early embryo.Crossref | GoogleScholarGoogle Scholar | 31022701PubMed |

Hansen, PJ (2020). The incompletely fulfilled promise of embryo transfer in cattle—why aren’t pregnancy rates greater and what can we do about it? Journal of Animal Science 98, skaa288.
The incompletely fulfilled promise of embryo transfer in cattle—why aren’t pregnancy rates greater and what can we do about it?Crossref | GoogleScholarGoogle Scholar | 33141879PubMed |

Hasan, MM, Viil, J, Lättekivi, F, Ord, J, Reshi, QUA, Jääger, K, Velthut-Meikas, A, Andronowska, A, Jaakma, Ü, Salumets, A, and Fazeli, A (2020). Bovine follicular fluid and extracellular vesicles derived from follicular fluid alter the bovine oviductal epithelial cells transcriptome. International Journal of Molecular Sciences 21, 5365.
Bovine follicular fluid and extracellular vesicles derived from follicular fluid alter the bovine oviductal epithelial cells transcriptome.Crossref | GoogleScholarGoogle Scholar |

Holt, WV, and Fazeli, A (2016). Sperm selection in the female mammalian reproductive tract. Focus on the oviduct: hypotheses, mechanisms, and new opportunities. Theriogenology 85, 105–112.
Sperm selection in the female mammalian reproductive tract. Focus on the oviduct: hypotheses, mechanisms, and new opportunities.Crossref | GoogleScholarGoogle Scholar | 26277704PubMed |

Howe, GR, and Black, DL (1963). Spermatozoan transport and leucocytic responses in the reproductive tract of calves. Reproduction 6, 305–311.
Spermatozoan transport and leucocytic responses in the reproductive tract of calves.Crossref | GoogleScholarGoogle Scholar |

Hunter RHF (1988) ‘The fallopian tubes: their role in fertility and infertility’. (Springer-Verlag: Berlin, Germany)
| Crossref |

Johnson, GP, English, A-M, Cronin, S, Hoey, DA, Meade, KG, and Fair, S (2017). Genomic identification, expression profiling, and functional characterization of CatSper channels in the bovine. Biology of Reproduction 97, 302–312.
Genomic identification, expression profiling, and functional characterization of CatSper channels in the bovine.Crossref | GoogleScholarGoogle Scholar | 29044427PubMed |

Kirichok, Y, and Lishko, PV (2011). Rediscovering sperm ion channels with the patch-clamp technique. Molecular Human Reproduction 17, 478–499.
Rediscovering sperm ion channels with the patch-clamp technique.Crossref | GoogleScholarGoogle Scholar | 21642646PubMed |

Kölle, S, Dubielzig, S, Reese, S, Wehrend, A, König, P, and Kummer, W (2009). Ciliary transport, gamete interaction, and effects of the early embryo in the oviduct: ex vivo analyses using a new digital videomicroscopic system in the cow. Biology of Reproduction 81, 267–274.
Ciliary transport, gamete interaction, and effects of the early embryo in the oviduct: ex vivo analyses using a new digital videomicroscopic system in the cow.Crossref | GoogleScholarGoogle Scholar | 19299315PubMed |

Konoshenko, MY, Lekchnov, EA, Vlassov, AV, and Laktionov, PP (2018). Isolation of extracellular vesicles: general methodologies and latest trends. BioMed Research International 2018, 8545347.
Isolation of extracellular vesicles: general methodologies and latest trends.Crossref | GoogleScholarGoogle Scholar | 29662902PubMed |

Kowalik, MK, Slonina, D, Rekawiecki, R, and Kotwica, J (2013). Expression of progesterone receptor membrane component (PGRMC) 1 and 2, serpine mRNA binding protein 1 (SERBP1) and nuclear progesterone receptor (PGR) in the bovine endometrium during the estrous cycle and the first trimester of pregnancy. Reproductive Biology 13, 15–23.
Expression of progesterone receptor membrane component (PGRMC) 1 and 2, serpine mRNA binding protein 1 (SERBP1) and nuclear progesterone receptor (PGR) in the bovine endometrium during the estrous cycle and the first trimester of pregnancy.Crossref | GoogleScholarGoogle Scholar | 23522067PubMed |

Kurian, NK, and Modi, D (2019). Extracellular vesicle mediated embryo-endometrial cross talk during implantation and in pregnancy. Journal of Assisted Reproduction and Genetics 36, 189–198.
Extracellular vesicle mediated embryo-endometrial cross talk during implantation and in pregnancy.Crossref | GoogleScholarGoogle Scholar | 30362057PubMed |

Lamy, J, Liere, P, Pianos, A, Aprahamian, F, Mermillod, P, and Saint-Dizier, M (2016). Steroid hormones in bovine oviductal fluid during the estrous cycle. Theriogenology 86, 1409–1420.
Steroid hormones in bovine oviductal fluid during the estrous cycle.Crossref | GoogleScholarGoogle Scholar | 27262884PubMed |

Lange-Consiglio, A, Perrini, C, Albini, G, Modina, S, Lodde, V, Orsini, E, Esposti, P, and Cremonesi, F (2017). Oviductal microvesicles and their effect on in vitro maturation of canine oocytes. Reproduction 154, 167–180.
Oviductal microvesicles and their effect on in vitro maturation of canine oocytes.Crossref | GoogleScholarGoogle Scholar | 28652254PubMed |

Leal, C, Cañon-Beltrán, K, Cajas, Y, Gallego, P, Beltrán-Breña, P, Hamdi, M, González, M, and Rizos, D (2020). 76 Extracellular vesicles from oviduct and uterus in sequential culture improve the quality of bovine embryos produced in vitro. Reproduction, Fertility and Development 32, 164–164.
76 Extracellular vesicles from oviduct and uterus in sequential culture improve the quality of bovine embryos produced in vitro.Crossref | GoogleScholarGoogle Scholar |

Leal, CLV, Cañón-Beltrán, K, Cajas, YN, Yaryes, A, Beltrán-Breña, P, Hamdi, M, Gutiérrez-Adán, A, González, ME, and Rizos, D (2021). 52 Extracellular vesicles from oviduct and uterus in sequential in vitro culture affects mitochondrial activity and lipid metabolism transcripts in bovine embryos. Reproduction, Fertility and Development 33, 133.
52 Extracellular vesicles from oviduct and uterus in sequential in vitro culture affects mitochondrial activity and lipid metabolism transcripts in bovine embryos.Crossref | GoogleScholarGoogle Scholar |

Lee, K-F, Yao, Y-Q, Kwok, K-L, Xu, J-S, and Yeung, WSB (2002). Early developing embryos affect the gene expression patterns in the mouse oviduct. Biochemical and Biophysical Research Communications 292, 564–570.
Early developing embryos affect the gene expression patterns in the mouse oviduct.Crossref | GoogleScholarGoogle Scholar | 11906198PubMed |

Lee, Y-L, Lee, K-F, Xu, J-S, He, Q-Y, Chiu, J-F, Lee, WM, Luk, JM, and Yeung, WSB (2004). The embryotrophic activity of oviductal cell-derived complement C3b and iC3b, a novel function of complement protein in reproduction. Journal of Biological Chemistry 279, 12763–12768.
The embryotrophic activity of oviductal cell-derived complement C3b and iC3b, a novel function of complement protein in reproduction.Crossref | GoogleScholarGoogle Scholar |

Leese, HJ, Hugentobler, SA, Gray, SM, Morris, DG, Sturmey, RG, Whitear, S-L, and Sreenan, JM (2008). Female reproductive tract fluids: composition, mechanism of formation and potential role in the developmental origins of health and disease. Reproduction, Fertility and Development 20, 1–8.
Female reproductive tract fluids: composition, mechanism of formation and potential role in the developmental origins of health and disease.Crossref | GoogleScholarGoogle Scholar |

Lefebvre, R, Lo, MC, and Suarez, SS (1997). Bovine sperm binding to oviductal epithelium involves fucose recognition. Biology of Reproduction 56, 1198–1204.
Bovine sperm binding to oviductal epithelium involves fucose recognition.Crossref | GoogleScholarGoogle Scholar | 9160719PubMed |

Lin, X, Pavani, KC, Smits, K, Deforce, D, Heindryckx, B, Van Soom, A, and Peelman, L (2019). Bta-miR-10b secreted by bovine embryos negatively impacts preimplantation embryo quality. Frontiers in Genetics 10, 757.
Bta-miR-10b secreted by bovine embryos negatively impacts preimplantation embryo quality.Crossref | GoogleScholarGoogle Scholar | 31507632PubMed |

Lloyd, RE, Romar, R, Matás, C, Gutiérrez-Adán, A, Holt, WV, and Coy, P (2009). Effects of oviductal fluid on the development, quality, and gene expression of porcine blastocysts produced in vitro. Reproduction 137, 679–687.
Effects of oviductal fluid on the development, quality, and gene expression of porcine blastocysts produced in vitro.Crossref | GoogleScholarGoogle Scholar | 19153191PubMed |

Lonergan, P (2007). State-of-the-art embryo technologies in cattle. Reproduction in Domestic Ruminants 64, 315–325.
State-of-the-art embryo technologies in cattle.Crossref | GoogleScholarGoogle Scholar |

Lonergan, P (2011). Influence of progesterone on oocyte quality and embryo development in cows. Theriogenology 76, 1594–1601.
Influence of progesterone on oocyte quality and embryo development in cows.Crossref | GoogleScholarGoogle Scholar | 21855985PubMed |

Lonergan, P, Fair, T, Forde, N, and Rizos, D (2016). Embryo development in dairy cattle. Theriogenology 86, 270–277.
Embryo development in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 27158131PubMed |

Lonergan, P, Rizos, D, Gutierrez-Adán, A, Moreira, PM, Pintado, B, de la Fuente, J, and Boland, MP (2003a). Temporal divergence in the pattern of messenger RNA expression in bovine embryos cultured from the zygote to blastocyst stage in vitro or in vivo. Biology of Reproduction 69, 1424–1431.
Temporal divergence in the pattern of messenger RNA expression in bovine embryos cultured from the zygote to blastocyst stage in vitro or in vivo.Crossref | GoogleScholarGoogle Scholar | 12826577PubMed |

Lonergan, P, Rizos, D, Kanka, J, Nemcova, L, Mbaye, AM, Kingston, M, Wade, M, Duffy, P, and Boland, MP (2003b). Temporal sensitivity of bovine embryos to culture environment after fertilization and the implications for blastocyst quality. Reproduction 126, 337–346.
Temporal sensitivity of bovine embryos to culture environment after fertilization and the implications for blastocyst quality.Crossref | GoogleScholarGoogle Scholar | 12968941PubMed |

Lopera-Vasquez, R, Hamdi, M, Maillo, V, Gutierrez-Adan, A, Bermejo-Alvarez, P, Ramírez, MÁ, Yáñez-Mó, M, and Rizos, D (2017a). Effect of bovine oviductal extracellular vesicles on embryo development and quality in vitro. Reproduction 153, 461–470.
Effect of bovine oviductal extracellular vesicles on embryo development and quality in vitro.Crossref | GoogleScholarGoogle Scholar | 28104825PubMed |

Lopera-Vasquez, R, Hamdi, M, Maillo, V, Lloreda, V, Coy, P, Gutierrez-Adan, A, Bermejo-Alvarez, P, and Rizos, D (2017b). Effect of bovine oviductal fluid on development and quality of bovine embryos produced in vitro. Reproduction, Fertility and Development 29, 621–629.
Effect of bovine oviductal fluid on development and quality of bovine embryos produced in vitro.Crossref | GoogleScholarGoogle Scholar |

Lopera-Vásquez, R, Hamdi, M, Fernandez-Fuertes, B, Maillo, V, Beltrán-Breña, P, Calle, A, Redruello, A, López-Martín, S, Gutierrez-Adán, A, Yañez-Mó, M, Ramirez, MÁ, and Rizos, D (2016). Extracellular vesicles from BOEC in in vitro embryo development and quality. PLoS ONE 11, e0148083.
Extracellular vesicles from BOEC in in vitro embryo development and quality.Crossref | GoogleScholarGoogle Scholar | 26845570PubMed |

Lösel, RM, Besong, D, Peluso, JJ, and Wehling, M (2008). Progesterone receptor membrane component 1—many tasks for a versatile protein. Steroids 73, 929–934.
Progesterone receptor membrane component 1—many tasks for a versatile protein.Crossref | GoogleScholarGoogle Scholar | 18249431PubMed |

Maillo, V, de Frutos, C, O’Gaora, P, Forde, N, Burns, GW, Spencer, TE, Gutierrez-Adan, A, Lonergan, P, and Rizos, D (2016). Spatial differences in gene expression in the bovine oviduct. Reproduction 152, 37–46.
Spatial differences in gene expression in the bovine oviduct.Crossref | GoogleScholarGoogle Scholar | 27069007PubMed |

Maillo, V, Gaora, PÓ, Forde, N, Besenfelder, U, Havlicek, V, Burns, GW, Spencer, TE, Gutierrez-Adan, A, Lonergan, P, and Rizos, D (2015). Oviduct-embryo interactions in cattle: two-way traffic or a one-way street? Biology of Reproduction 92, 144.
Oviduct-embryo interactions in cattle: two-way traffic or a one-way street?Crossref | GoogleScholarGoogle Scholar | 25926440PubMed |

Marey, MA, Liu, J, Kowsar, R, Haneda, S, Matsui, M, Sasaki, M, Shimizu, T, Hayakawa, H, Wijayagunawardane, MPB, Hussein, FM, and Miyamoto, A (2014). Bovine oviduct epithelial cells downregulate phagocytosis of sperm by neutrophils: prostaglandin E2 as a major physiological regulator. Reproduction 147, 211–219.
Bovine oviduct epithelial cells downregulate phagocytosis of sperm by neutrophils: prostaglandin E2 as a major physiological regulator.Crossref | GoogleScholarGoogle Scholar | 24255155PubMed | 24255155PubMed |

Marey, MA, Matsukawa, H, Sasaki, M, Ezz, MA, Yousef, MS, Takahashi, K-I, and Miyamoto, A (2020). Bovine oviduct epithelial cells suppress the phagocytic activity of neutrophils towards sperm but not for bacteria in vitro: Immunofluorescence and electron microscopic observations. Histology and Histopathology 35, 589–597.
Bovine oviduct epithelial cells suppress the phagocytic activity of neutrophils towards sperm but not for bacteria in vitro: Immunofluorescence and electron microscopic observations.Crossref | GoogleScholarGoogle Scholar | 31621887PubMed | 31621887PubMed |

Mathew, DJ, Sánchez, JM, Passaro, C, Charpigny, G, Behura, SK, Spencer, TE, and Lonergan, P (2019). Interferon tau-dependent and independent effects of the bovine conceptus on the endometrial transcriptome. Biology of Reproduction 100, 365–380.
Interferon tau-dependent and independent effects of the bovine conceptus on the endometrial transcriptome.Crossref | GoogleScholarGoogle Scholar | 30203055PubMed | 30203055PubMed |

Mazzarella, R, Bastos, NM, Bridi, A, del Collado, M, Andrade, GM, Pinzon, J, Prado, CM, Silva, LA, Meirelles, FV, Pugliesi, G, Perecin, F, and da Silveira, JC (2021). Changes in oviductal cells and small extracellular vesicles miRNAs in pregnant cows. Frontiers in Veterinary Science 8, 639752.
Changes in oviductal cells and small extracellular vesicles miRNAs in pregnant cows.Crossref | GoogleScholarGoogle Scholar | 33748215PubMed | 33748215PubMed |

Mellisho, EA, Briones, MA, Velásquez, AE, Cabezas, J, Castro, FO, and Rodríguez-Álvarez, L (2019). Extracellular vesicles secreted during blastulation show viability of bovine embryos. Reproduction 158, 477–492.
Extracellular vesicles secreted during blastulation show viability of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 31600718PubMed | 31600718PubMed |

Mellisho, EA, Velásquez, AE, Nuñez, MJ, Cabezas, JG, Cueto, JA, Fader, C, Castro, FO, and Rodríguez-Álvarez, L (2017). Identification and characteristics of extracellular vesicles from bovine blastocysts produced in vitro. PLoS ONE 12, e0178306.
Identification and characteristics of extracellular vesicles from bovine blastocysts produced in vitro.Crossref | GoogleScholarGoogle Scholar | 28542562PubMed | 28542562PubMed |

Melo-Baez, B, Wong, YS, Aguilera, CJ, Cabezas, J, Mançanares, ACF, Riadi, G, Castro, FO, and Rodriguez-Alvarez, L (2020). MicroRNAs from extracellular vesicles secreted by bovine embryos as early biomarkers of developmental competence. International Journal of Molecular Sciences 21, 8888.
MicroRNAs from extracellular vesicles secreted by bovine embryos as early biomarkers of developmental competence.Crossref | GoogleScholarGoogle Scholar |

Mondéjar, I, Martínez-Martínez, I, Avilés, M, and Coy, P (2013). Identification of potential oviductal factors responsible for zona pellucida hardening and monospermy during fertilization in mammals. Biology of Reproduction 89, 67.
Identification of potential oviductal factors responsible for zona pellucida hardening and monospermy during fertilization in mammals.Crossref | GoogleScholarGoogle Scholar | 23863406PubMed | 23863406PubMed |

Mullen, MP, Elia, G, Hilliard, M, Parr, MH, Diskin, MG, Evans, ACO, and Crowe, MA (2012). Proteomic characterization of histotroph during the preimplantation phase of the estrous cycle in cattle. Journal of Proteome Research 11, 3004–3018.
Proteomic characterization of histotroph during the preimplantation phase of the estrous cycle in cattle.Crossref | GoogleScholarGoogle Scholar | 22463384PubMed | 22463384PubMed |

Muñoz, M, Corrales, FJ, Caamaño, JN, Díez, C, Trigal, B, Mora, MI, Martín, D, Carrocera, S, and Gómez, E (2012). Proteome of the early embryo–maternal dialogue in the cattle uterus. Journal of Proteome Research 11, 751–766.
Proteome of the early embryo–maternal dialogue in the cattle uterus.Crossref | GoogleScholarGoogle Scholar | 22148898PubMed | 22148898PubMed |

Niemann, H, and Wrenzycki, C (2000). Alterations of expression of developmentally important genes in preimplantation bovine embryos by in vitro culture conditions: implications for subsequent development. Theriogenology 53, 21–34.
Alterations of expression of developmentally important genes in preimplantation bovine embryos by in vitro culture conditions: implications for subsequent development.Crossref | GoogleScholarGoogle Scholar | 10735059PubMed | 10735059PubMed |

Nordberg, J, and Arnér, ESJ (2001). Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radical Biology and Medicine 31, 1287–1312.
Reactive oxygen species, antioxidants, and the mammalian thioredoxin system.Crossref | GoogleScholarGoogle Scholar | 11728801PubMed | 11728801PubMed |

Okada, H, Hirose, Y, Manonmani, P, Uda, A, Ito, M, and Sankai, T (2005). Characterization of an immortalized oviduct cell line from the cynomolgus monkey (Macaca fascicularis. Journal of Medical Primatology 34, 67–72.
Characterization of an immortalized oviduct cell line from the cynomolgus monkey (Macaca fascicularis.Crossref | GoogleScholarGoogle Scholar | 15860112PubMed | 15860112PubMed |

Ortiz, ME, Bedregal, P, Carvajal, MI, and Croxatto, HB (1986). Fertilized and unfertilized ova are transported at different rates by the hamster oviduct. Biology of Reproduction 34, 777–781.
Fertilized and unfertilized ova are transported at different rates by the hamster oviduct.Crossref | GoogleScholarGoogle Scholar | 3708057PubMed | 3708057PubMed |

Ortiz, ME, Llados, C, and Croxatto, HB (1989). Embryos of different ages transferred to the rat oviduct enter the uterus at different times. Biology of Reproduction 41, 381–384.
Embryos of different ages transferred to the rat oviduct enter the uterus at different times.Crossref | GoogleScholarGoogle Scholar | 2590709PubMed | 2590709PubMed |

Passaro, C, Tutt, D, Bagés-Arnal, S, Maicas, C, Laguna-Barraza, R, Gutierrez-Adán, A, Browne, JA, Rath, D, Behura, SK, Spencer, TE, Fair, T, and Lonergan, P (2019). Global transcriptomic response of bovine endometrium to blastocyst-stage embryos. Reproduction 158, 223–235.
Global transcriptomic response of bovine endometrium to blastocyst-stage embryos.Crossref | GoogleScholarGoogle Scholar | 31247587PubMed | 31247587PubMed |

Passaro, C, Tutt, D, Mathew, DJ, Sanchez, JM, Browne, JA, Boe-Hansen, GB, Fair, T, and Lonergan, P (2018). Blastocyst-induced changes in the bovine endometrial transcriptome. Reproduction 156, 219–229.
Blastocyst-induced changes in the bovine endometrial transcriptome.Crossref | GoogleScholarGoogle Scholar | 30021913PubMed | 30021913PubMed |

Pavani, KC, Hendrix, A, Van Den Broeck, W, Couck, L, Szymanska, K, Lin, X, De Koster, J, Van Soom, A, and Leemans, B (2019). Isolation and characterization of functionally active extracellular vesicles from culture medium conditioned by bovine embryos in vitro. International Journal of Molecular Sciences 20, 38.
Isolation and characterization of functionally active extracellular vesicles from culture medium conditioned by bovine embryos in vitro.Crossref | GoogleScholarGoogle Scholar |

Pérez-Cerezales, S, Boryshpolets, S, Afanzar, O, Brandis, A, Nevo, R, Kiss, V, and Eisenbach, M (2015). Involvement of opsins in mammalian sperm thermotaxis. Scientific Reports 5, 16146.
Involvement of opsins in mammalian sperm thermotaxis.Crossref | GoogleScholarGoogle Scholar | 26537127PubMed | 26537127PubMed |

Pillai, VV, Weber, DM, Phinney, BS, and Selvaraj, V (2017). Profiling of proteins secreted in the bovine oviduct reveals diverse functions of this luminal microenvironment. PLoS ONE 12, e0188105.
Profiling of proteins secreted in the bovine oviduct reveals diverse functions of this luminal microenvironment.Crossref | GoogleScholarGoogle Scholar | 29155854PubMed | 29155854PubMed |

Pontes, JHF, Nonato-Junior, I, Sanches, BV, Ereno-Junior, JC, Uvo, S, Barreiros, TRR, Oliveira, JA, Hasler, JF, and Seneda, MM (2009). Comparison of embryo yield and pregnancy rate between in vivo and in vitro methods in the same Nelore (Bos indicus) donor cows. Theriogenology 71, 690–697.
Comparison of embryo yield and pregnancy rate between in vivo and in vitro methods in the same Nelore (Bos indicus) donor cows.Crossref | GoogleScholarGoogle Scholar |

Qamar, AY, Mahiddine, FY, Bang, S, Fang, X, Shin, ST, Kim, MJ, and Cho, J (2020). Extracellular vesicle mediated crosstalk between the gametes, conceptus, and female reproductive tract. Frontiers in Veterinary Science 7, .
Extracellular vesicle mediated crosstalk between the gametes, conceptus, and female reproductive tract.Crossref | GoogleScholarGoogle Scholar | 33195625PubMed | 33195625PubMed |

Qu, P, Qing, S, Liu, R, Qin, H, Wang, W, Qiao, F, Ge, H, Liu, J, Zhang, Y, Cui, W, and Wang, Y (2017). Effects of embryo-derived exosomes on the development of bovine cloned embryos. PLoS ONE 12, e0174535.
Effects of embryo-derived exosomes on the development of bovine cloned embryos.Crossref | GoogleScholarGoogle Scholar | 28350875PubMed | 28350875PubMed |

Qu, P, Zhao, Y, Wang, R, Zhang, Y, Li, L, Fan, J, and Liu, E (2019). Extracellular vesicles derived from donor oviduct fluid improved birth rates after embryo transfer in mice. Reproduction, Fertility and Development 31, 324–332.
Extracellular vesicles derived from donor oviduct fluid improved birth rates after embryo transfer in mice.Crossref | GoogleScholarGoogle Scholar |

Ramos-Ibeas, P, Calle, A, Pericuesta, E, Laguna-Barraza, R, Moros-Mora, R, Lopera-Vásquez, R, Maillo, V, Yáñez-Mó, M, Gutiérrez-Adán, A, Rizos, D, and Ramírez, MÁ (2014). An efficient system to establish biopsy-derived trophoblastic cell lines from bovine embryos. Biology of Reproduction 91, 15.
An efficient system to establish biopsy-derived trophoblastic cell lines from bovine embryos.Crossref | GoogleScholarGoogle Scholar | 24855108PubMed | 24855108PubMed |

Rashid, MB, Talukder, AK, Kusama, K, Haneda, S, Takedomi, T, Yoshino, H, Moriyasu, S, Matsui, M, Shimada, M, Imakawa, K, and Miyamoto, A (2018). Evidence that interferon-tau secreted from Day-7 embryo in vivo generates anti-inflammatory immune response in the bovine uterus. Biochemical and Biophysical Research Communications 500, 879–884.
Evidence that interferon-tau secreted from Day-7 embryo in vivo generates anti-inflammatory immune response in the bovine uterus.Crossref | GoogleScholarGoogle Scholar | 29702095PubMed | 29702095PubMed |

Rath, D, Schuberth, HJ, Coy, P, and Taylor, U (2008). Sperm interactions from insemination to fertilization. Reproduction in Domestic Animals 43, 2–11.
Sperm interactions from insemination to fertilization.Crossref | GoogleScholarGoogle Scholar | 19068027PubMed | 19068027PubMed |

Reinhart, KC, Dubey, RK, Cometti, B, Keller, PJ, and Rosselli, M (2003). Differential effects of natural and environmental estrogens on endothelin synthesis in bovine oviduct cells. Biology of Reproduction 68, 1430–1436.
Differential effects of natural and environmental estrogens on endothelin synthesis in bovine oviduct cells.Crossref | GoogleScholarGoogle Scholar | 12606437PubMed | 12606437PubMed |

Rizos, D, Clemente, M, Bermejo-Alvarez, P, Fuente, JDL, Lonergan, P, and Gutiérrez-Adán, A (2008). Consequences of in vitro culture conditions on embryo development and quality. Reproduction in Domestic Animals 43, 44–50.
Consequences of in vitro culture conditions on embryo development and quality.Crossref | GoogleScholarGoogle Scholar | 18803756PubMed | 18803756PubMed |

Rizos, D, Fair, T, Papadopoulos, S, Boland, MP, and Lonergan, P (2002a). Developmental, qualitative, and ultrastructural differences between ovine and bovine embryos produced in vivo or in vitro. Molecular Reproduction and Development 62, 320–327.
Developmental, qualitative, and ultrastructural differences between ovine and bovine embryos produced in vivo or in vitro.Crossref | GoogleScholarGoogle Scholar | 12112595PubMed | 12112595PubMed |

Rizos, D, Pintado, B, de la Fuente, J, Lonergan, P, and Gutiérrez-Adán, A (2007). Development and pattern of mRNA relative abundance of bovine embryos cultured in the isolated mouse oviduct in organ culture. Molecular Reproduction and Development 74, 716–723.
Development and pattern of mRNA relative abundance of bovine embryos cultured in the isolated mouse oviduct in organ culture.Crossref | GoogleScholarGoogle Scholar | 17154298PubMed | 17154298PubMed |

Rizos, D, Ward, F, Duffy, P, Boland, MP, and Lonergan, P (2002b). Consequences of bovine oocyte maturation, fertilization or early embryo development in vitro versus in vivo: implications for blastocyst yield and blastocyst quality. Molecular Reproduction and Development 61, 234–248.
Consequences of bovine oocyte maturation, fertilization or early embryo development in vitro versus in vivo: implications for blastocyst yield and blastocyst quality.Crossref | GoogleScholarGoogle Scholar | 11803560PubMed | 11803560PubMed |

Rødgaard, T, Heegaard, PMH, and Callesen, H (2015). Non-invasive assessment of in-vitro embryo quality to improve transfer success. Reproductive BioMedicine Online 31, 585–592.
Non-invasive assessment of in-vitro embryo quality to improve transfer success.Crossref | GoogleScholarGoogle Scholar | 26380864PubMed | 26380864PubMed |

Rodríguez-Alonso, B, Hamdi, M, Sánchez, JM, Maillo, V, Gutierrez-Adan, A, Lonergan, P, and Rizos, D (2019). An approach to study the local embryo effect on gene expression in the bovine oviduct epithelium in vivo. Reproduction in Domestic Animals 54, 1516–1523.
An approach to study the local embryo effect on gene expression in the bovine oviduct epithelium in vivo.Crossref | GoogleScholarGoogle Scholar | 31472078PubMed | 31472078PubMed |

Rodríguez-Alonso, B, Sánchez, JM, Hamdi, M, McDonald, M, Havlicek, V, Besenfelder, U, Lonergan, P, and Rizos, D (2020). Asynchrony between the early embryo and the reproductive tract affects subsequent embryo development in cattle. Reproduction, Fertility and Development 32, 564–571.
Asynchrony between the early embryo and the reproductive tract affects subsequent embryo development in cattle.Crossref | GoogleScholarGoogle Scholar |

Saint-Dizier, M, Schoen, J, Chen, S, Banliat, C, and Mermillod, P (2019). Composing the early embryonic microenvironment: physiology and regulation of oviductal secretions. International Journal of Molecular Sciences 21, 223.
Composing the early embryonic microenvironment: physiology and regulation of oviductal secretions.Crossref | GoogleScholarGoogle Scholar |

Salilew-Wondim, D, Fournier, E, Hoelker, M, Saeed-Zidane, M, Tholen, E, Looft, C, Neuhoff, C, Besenfelder, U, Havlicek, V, Rings, F, Gagné, D, Sirard, M-A, Robert, C, Shojaei Saadi, HA, Gad, A, Schellander, K, and Tesfaye, D (2015). Genome-wide DNA methylation patterns of bovine blastocysts developed in vivo from embryos completed different stages of development in vitro. PLoS ONE 10, e0140467.
Genome-wide DNA methylation patterns of bovine blastocysts developed in vivo from embryos completed different stages of development in vitro.Crossref | GoogleScholarGoogle Scholar | 26536655PubMed | 26536655PubMed |

Salilew-Wondim, D, Gebremedhn, S, Hoelker, M, Tholen, E, Hailay, T, and Tesfaye, D (2020). The role of microRNAs in mammalian fertility: from gametogenesis to embryo implantation. International Journal of Molecular Sciences 21, 585.
The role of microRNAs in mammalian fertility: from gametogenesis to embryo implantation.Crossref | GoogleScholarGoogle Scholar |

Sánchez, JM, Mathew, DJ, Behura, SK, Passaro, C, Charpigny, G, Butler, ST, Spencer, TE, and Lonergan, P (2019a). Bovine endometrium responds differentially to age-matched short and long conceptuses. Biology of Reproduction 101, 26–39.
Bovine endometrium responds differentially to age-matched short and long conceptuses.Crossref | GoogleScholarGoogle Scholar | 30977805PubMed | 30977805PubMed |

Sánchez, JM, Mathew, DJ, Passaro, C, Fair, T, and Lonergan, P (2018). Embryonic maternal interaction in cattle and its relationship with fertility. Reproduction in Domestic Animals 53, 20–27.
Embryonic maternal interaction in cattle and its relationship with fertility.Crossref | GoogleScholarGoogle Scholar | 30238655PubMed | 30238655PubMed |

Sánchez, JM, Simintiras, CA, and Lonergan, P (2019b). Aspects of embryo–maternal communication in establishment of pregnancy in cattle. Animal Reproduction 16, 376–385.
Aspects of embryo–maternal communication in establishment of pregnancy in cattle.Crossref | GoogleScholarGoogle Scholar | 32435281PubMed | 32435281PubMed |

Sartori, R, Bastos, MR, and Wiltbank, MC (2010). Factors affecting fertilisation and early embryo quality in single- and superovulated dairy cattle. Reproduction, Fertility and Development 22, 151–158.
Factors affecting fertilisation and early embryo quality in single- and superovulated dairy cattle.Crossref | GoogleScholarGoogle Scholar |

Schmaltz-Panneau, B, Cordova, A, Dhorne-Pollet, S, Hennequet-Antier, C, Uzbekova, S, Martinot, E, Doret, S, Martin, P, Mermillod, P, and Locatelli, Y (2014). Early bovine embryos regulate oviduct epithelial cell gene expression during in vitro co-culture. Animal Reproduction Science 149, 103–116.
Early bovine embryos regulate oviduct epithelial cell gene expression during in vitro co-culture.Crossref | GoogleScholarGoogle Scholar | 25113901PubMed | 25113901PubMed |

Senger PL (2003) ‘Pathways to pregnancy and parturition’. (Current Conceptions: Pullman, WA, USA)

Smits, K, De Coninck, DIM, Van Nieuwerburgh, F, Govaere, J, Van Poucke, M, Peelman, L, Deforce, D, and Van Soom, A (2016). The equine embryo influences immune-related gene expression in the oviduct. Biology of Reproduction 94, 36.
The equine embryo influences immune-related gene expression in the oviduct.Crossref | GoogleScholarGoogle Scholar | 26740593PubMed | 26740593PubMed |

Spencer, TE (2014). Biological roles of uterine glands in pregnancy. Seminars in Reproductive Medicine 32, 346–357.
Biological roles of uterine glands in pregnancy.Crossref | GoogleScholarGoogle Scholar | 24959816PubMed | 24959816PubMed |

Sponchiado, M, Gomes, NS, Fontes, PK, Martins, T, Del Collado, M, Pastore, AdA, Pugliesi, G, Nogueira, MFG, and Binelli, M (2017). Pre-hatching embryo-dependent and -independent programming of endometrial function in cattle. PLoS ONE 12, e0175954.
Pre-hatching embryo-dependent and -independent programming of endometrial function in cattle.Crossref | GoogleScholarGoogle Scholar | 28423001PubMed | 28423001PubMed |

Sponchiado, M, Gonella-Diaza, AM, Rocha, CC, Turco, EGL, Pugliesi, G, Leroy, JLMR, and Binelli, M (2019). The pre-hatching bovine embryo transforms the uterine luminal metabolite composition in vivo. Scientific Reports 9, 8354.
The pre-hatching bovine embryo transforms the uterine luminal metabolite composition in vivo.Crossref | GoogleScholarGoogle Scholar | 31175317PubMed | 31175317PubMed |

Sponchiado, M, Marei, WFA, Beemster, GTS, Bols, PEJ, Binelli, M, and Leroy, JLMR (2020). Molecular interactions at the bovine embryo–endometrial epithelium interface. Reproduction 160, 887–903.
Molecular interactions at the bovine embryo–endometrial epithelium interface.Crossref | GoogleScholarGoogle Scholar | 33112768PubMed | 33112768PubMed |

Suarez, SS (2008). Regulation of sperm storage and movement in the mammalian oviduct. International Journal of Developmental Biology 52, 455–462.
Regulation of sperm storage and movement in the mammalian oviduct.Crossref | GoogleScholarGoogle Scholar |

Suarez SS (2015) Gamete and zygote transport. In ‘Knobil and Neill’s physiology of reproduction’. pp. 197–232. (Elsevier: San Diego, CA, USA)
| Crossref |

Suarez, SS, Brockman, K, and Lefebvre, R (1997). Distribution of mucus and sperm in bovine oviducts after artificial insemination: the physical environment of the oviductal sperm reservoir. Biology of Reproduction 56, 447–453.
Distribution of mucus and sperm in bovine oviducts after artificial insemination: the physical environment of the oviductal sperm reservoir.Crossref | GoogleScholarGoogle Scholar | 9116145PubMed | 9116145PubMed |

Sutton, R, Nancarrow, CD, Wallace, AL, and Rigby, NW (1984). Identification of an oestrus-associated glycoprotein in oviducal fluid of the sheep. Journal of Reproduction Fertility 72, 415–422.
Identification of an oestrus-associated glycoprotein in oviducal fluid of the sheep.Crossref | GoogleScholarGoogle Scholar | 6542589PubMed | 6542589PubMed |

Takahashi, H, Haneda, S, Kayano, M, and Matsui, M (2016). Differences in progesterone concentrations and mRNA expressions of progesterone receptors in bovine endometrial tissue between the uterine horns ipsilateral and contralateral to the corpus luteum. The Journal of Veterinary Medical Science 78, 613–618.
Differences in progesterone concentrations and mRNA expressions of progesterone receptors in bovine endometrial tissue between the uterine horns ipsilateral and contralateral to the corpus luteum.Crossref | GoogleScholarGoogle Scholar | 26782011PubMed | 26782011PubMed |

Talukder, AK, Rashid, MB, Yousef, MS, Kusama, K, Shimizu, T, Shimada, M, Suarez, SS, Imakawa, K, and Miyamoto, A (2018). Oviduct epithelium induces interferon-tau in bovine Day-4 embryos, which generates an anti-inflammatory response in immune cells. Scientific Reports 8, 7850.
Oviduct epithelium induces interferon-tau in bovine Day-4 embryos, which generates an anti-inflammatory response in immune cells.Crossref | GoogleScholarGoogle Scholar | 29777205PubMed | 29777205PubMed |

Talukder, AK, Yousef, MS, Rashid, MB, Awai, K, Acosta, TJ, Shimizu, T, Okuda, K, Shimada, M, Imakawa, K, and Miyamoto, A (2017). Bovine embryo induces an anti-inflammatory response in uterine epithelial cells and immune cells in vitro: possible involvement of interferon tau as an intermediator. The Journal of Reproduction and Development 63, 425–434.
Bovine embryo induces an anti-inflammatory response in uterine epithelial cells and immune cells in vitro: possible involvement of interferon tau as an intermediator.Crossref | GoogleScholarGoogle Scholar | 28603222PubMed | 28603222PubMed |

Tesfaye, D, Salilew-Wondim, D, Gebremedhn, S, Sohel, M, Pandey, H, Hoelker, M, and Schellander, K (2016). Potential role of microRNAs in mammalian female fertility. Reproduction, Fertility and Development 29, 8–23.
Potential role of microRNAs in mammalian female fertility.Crossref | GoogleScholarGoogle Scholar |

Théry, C, Witwer, KW, Aikawa, E, Alcaraz, MJ, Anderson, JD, Andriantsitohaina, R, Antoniou, A, Arab, T, Archer, F, Atkin-Smith, GK, et al. (2018). Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles 7, 1535750.
Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines.Crossref | GoogleScholarGoogle Scholar | 30637094PubMed | 30637094PubMed |

Töpfer-Petersen, E, Wagner, A, Friedrich, J, Petrunkina, A, Ekhlasi-Hundrieser, M, Waberski, D, and Drommer, W (2002). Function of the mammalian oviductal sperm reservoir. Journal of Experimental Zoology 292, 210–215.
Function of the mammalian oviductal sperm reservoir.Crossref | GoogleScholarGoogle Scholar |

Tung, CK, Hu, L, Fiore, AG, Ardon, F, Hickman, DG, Gilbert, RO, Suarez, SS, and Wu, M (2015). Microgrooves and fluid flows provide preferential passageways for sperm over pathogen Tritrichomonas foetus. Proceedings of the National Academy of Sciences of the United States of America 112, 5431–5436.
Microgrooves and fluid flows provide preferential passageways for sperm over pathogen Tritrichomonas foetus.Crossref | GoogleScholarGoogle Scholar | 25870286PubMed | 25870286PubMed |

Urrego, R, Rodriguez-Osorio, N, and Niemann, H (2014). Epigenetic disorders and altered gene expression after use of assisted reproductive technologies in domestic cattle. Epigenetics 9, 803–815.
Epigenetic disorders and altered gene expression after use of assisted reproductive technologies in domestic cattle.Crossref | GoogleScholarGoogle Scholar | 24709985PubMed | 24709985PubMed |

Vajta, G, Rienzi, L, Cobo, A, and Yovich, J (2010). Embryo culture: can we perform better than nature? Reproductive BioMedicine Online 20, 453–469.
Embryo culture: can we perform better than nature?Crossref | GoogleScholarGoogle Scholar | 20202911PubMed | 20202911PubMed |

van Niekerk, CH, and Gerneke, WH (1966). Persistence and parthenogentic cleavage of tubal ova in the mare. Onderstepoort Journal of Veterinary Research 33, 195–232.

van Niel, G, D’Angelo, G, and Raposo, G (2018). Shedding light on the cell biology of extracellular vesicles. Nature Reviews Molecular Cell Biology 19, 213–228.
Shedding light on the cell biology of extracellular vesicles.Crossref | GoogleScholarGoogle Scholar | 29339798PubMed | 29339798PubMed |

Wijayagunawardane, MP, Miyamoto, A, Cerbito, WA, Acosta, TJ, Takagi, M, and Sato, K (1998). Local distributions of oviductal estradiol, progesterone, prostaglandins, oxytocin and endothelin-1 in the cyclic cow. Theriogenology 49, 607–618.
Local distributions of oviductal estradiol, progesterone, prostaglandins, oxytocin and endothelin-1 in the cyclic cow.Crossref | GoogleScholarGoogle Scholar | 10732039PubMed | 10732039PubMed |

Wiltbank, MC, Baez, GM, Garcia-Guerra, A, Toledo, MZ, Monteiro, PLJ, Melo, LF, Ochoa, JC, Santos, JEP, and Sartori, R (2016). Pivotal periods for pregnancy loss during the first trimester of gestation in lactating dairy cows. Theriogenology 86, 239–253.
Pivotal periods for pregnancy loss during the first trimester of gestation in lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 27238438PubMed | 27238438PubMed |

Witwer, KW, Buzás, EI, Bemis, LT, Bora, A, Lässer, C, Lötvall, J, Nolte-’t Hoen, EN, Piper, MG, Sivaraman, S, Skog, J, Théry, C, Wauben, MH, and Hochberg, F (2013). Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. Journal of Extracellular Vesicles 2, .
Standardization of sample collection, isolation and analysis methods in extracellular vesicle research.Crossref | GoogleScholarGoogle Scholar | 24009894PubMed | 24009894PubMed |

Wolf, E, Arnold, GJ, Bauersachs, S, Beier, HM, Blum, H, Einspanier, R, Fröhlich, T, Herrler, A, Hiendleder, S, Kölle, S, Prelle, K, Reichenbach, H-D, Stojkovic, M, Wenigerkind, H, and Sinowatz, F (2003). Embryo–maternal communication in bovine – strategies for deciphering a complex cross-talk. Reproduction in Domestic Animals 38, 276–289.
Embryo–maternal communication in bovine – strategies for deciphering a complex cross-talk.Crossref | GoogleScholarGoogle Scholar | 12887567PubMed | 12887567PubMed |

Wydooghe, E, Vandaele, L, Heras, S, De Sutter, P, Deforce, D, Peelman, L, De Schauwer, C, and Van Soom, A (2017). Autocrine embryotropins revisited: how do embryos communicate with each other in vitro when cultured in groups? Biological Reviews 92, 505–520.
Autocrine embryotropins revisited: how do embryos communicate with each other in vitro when cultured in groups?Crossref | GoogleScholarGoogle Scholar | 26608222PubMed | 26608222PubMed |

Yániz, JL, Lopez-Gatius, F, Santolaria, P, and Mullins, KJ (2000). Study of the functional anatomy of bovine oviductal mucosa. The Anatomical Record 260, 268–278.
Study of the functional anatomy of bovine oviductal mucosa.Crossref | GoogleScholarGoogle Scholar | 11066037PubMed | 11066037PubMed |

Yao, N, Wan, P-C, Hao, Z-D, Gao, F-F, Yang, L, Cui, M-S, Wu, Y, Liu, J-H, Liu, S, Chen, H, and Zeng, S-M (2009). Expression of interferon-tau mRNA in bovine embryos derived from different procedures. Reproduction in Domestic Animals 44, 132–139.
Expression of interferon-tau mRNA in bovine embryos derived from different procedures.Crossref | GoogleScholarGoogle Scholar | 19019066PubMed | 19019066PubMed |

Yousef, MS, Marey, MA, Hambruch, N, Hayakawa, H, Shimizu, T, Hussien, HA, Abdel-Razek, A-RK, Pfarrer, C, and Miyamoto, A (2016). Sperm binding to oviduct epithelial cells enhances TGFB1 and IL10 expressions in epithelial cells as well as neutrophils in vitro: prostaglandin E2 as a main regulator of anti-inflammatory response in the bovine oviduct. PLoS ONE 11, e0162309.
Sperm binding to oviduct epithelial cells enhances TGFB1 and IL10 expressions in epithelial cells as well as neutrophils in vitro: prostaglandin E2 as a main regulator of anti-inflammatory response in the bovine oviduct.Crossref | GoogleScholarGoogle Scholar | 27662642PubMed | 27662642PubMed |

Zhang, Y, Zhou, J, Li, M-Q, Xu, J, Zhang, J-P, and Jin, L-P (2019). MicroRNA-184 promotes apoptosis of trophoblast cells via targeting WIG1 and induces early spontaneous abortion. Cell Death and Disease 10, 223.
MicroRNA-184 promotes apoptosis of trophoblast cells via targeting WIG1 and induces early spontaneous abortion.Crossref | GoogleScholarGoogle Scholar | 30833572PubMed | 30833572PubMed |