Acessibilidade / Reportar erro

40 years of AETE: the contribution of scientists and practitioners to the progress of reproductive biotechnologies in Europe

Abstract

This conference celebrates the 40th anniversary of AETE. Over the past 40 years, AETE has served as a forum for scientists, practitioners, and students working in assisted animal reproduction in livestock species. AETE conferences have reflected developments in the field, from basic to applied science, as well as regulatory changes in assisted animal reproduction practices. Europe has led the way in these developments for many years, progressing from artificial insemination, embryo transfer, and cryopreservation to semen sexing, in vitro production of embryos, cloning by nuclear transfer, genomic selection, and the rescue of highly endangered species. These significant contributions were made possible by the support of funding agencies, both at the national and European levels, promoting cooperation between scientists and practitioners. Assisted reproduction, and animal breeding more generally, face opposition from various groups, including animal rights activists, vegetarians, proponents of organic farming, environmentalists, certain political parties, and increasing regulatory burdens. These challenges seriously affect funding for scientific research, the work of practitioners, and the breeding industry as a whole. It is crucial to invest time and resources in communication to remind the public, politicians, and regulators of the achievements in this field and the contributions made to the food supply chain and the care of the rural and natural environment.

Keywords:
assisted reproduction techniques; biotechnologies; AETE; Europe

Introduction

Europe has always been at the fore front and a leader in the development of assisted reproduction biotechnologies, more specifically termed ART (Assisted Reproduction Technologies). Lazzaro Spallanzani (Italy, 1729-1799) was the first to perform the first successful artificial insemination in a bitch (Lonergan, 2018Lonergan P. Review: historical and futuristic developments in bovine semen technology. Animal. 2018;12(s1):s4-s18. http://doi.org/10.1017/S175173111800071X. PMid:29642968.
http://doi.org/10.1017/S175173111800071X...
) and Walter Heape (United Kingdom,1855-1929) was the first to perform embryo transfer (Betteridge, 2003Betteridge KJ. A history of farm animal embryo transfer and some associated techniques. Anim Reprod Sci. 2003;79(3-4):203-44. http://doi.org/10.1016/S0378-4320(03)00166-0 PMid:14643106.
http://doi.org/10.1016/S0378-4320(03)001...
). Artificial insemination and embryo transfer were, and still are, the cornerstones of reproductive biotechnologies in mammalian reproduction. Following our forebears, many pioneers, both in academia and in practice, stepped in to bring reproductive biotechnologies to what we know and practice today, as well as opening new windows for younger generations to look into the future. This paper will not pretend to provide a complete and referenced review of 40 years of reproductive biotechnologies in Europe; rather, it offers a personal perspective. Having worked for 40 years both in academia and in industry, I will discuss on how science and practice have fed each other, primarily through scientific societies like AETE (Association of Embryo Technology in Europe), whose 40th anniversary we are celebrating this year.

Scientific societies like AETE in Europe, or IETS (International Embryo Technology Society) internationally, as well as national societies, have played an important role in advancing the field by bringing together scientists and practitioners around the table to discuss findings in research laboratories, the needs of the industry and practitioners and, most importantly, creating a forum for students to present their work and network with the community during 2 to 3 days meetings, that always included social events to facilitate this. The never-ending struggle of the Board of Governors of these societies and the program chairs has always been, and still is, to find the right balance to attract both scientists and practitioners. Moreover, an important part of such meetings are the exhibitors, not only because they provided the sponsorship to pay part of the expenses, but primarily to showcase newly developed tools, consumables, disposable and reagents required as the procedures developed and the regulatory requirements dictated.

Forty years ago, much of the work presented and discussed was related with cattle, but over the years, reproductive biotechnologies have widened their application to include other livestock species like small ruminants, pigs, buffaloes, horses, etc. but also endangered and exotic species, genomics selection, stem cells and genome editing.

In the paper celebrating the 30th year of AETE (Thibier, 2014Thibier M. The European embryo transfer industry in cattle - a challange in 1984 - a success in 2014 -and well supported and reported by the AETE. In: Proceedings of the 30th Scientific meeting of A.E.T.E.; 2014 Sept 12-13; Dresden, Germany. Dresden: A.E.T.E.; 2014. p. 1-13.) there is a detailed narration of the birth and evolution of AETE. In this paper I will attempt to outline what European scientists and practitioners have contributed to the advancement of reproductive biotechnologies, not only in Europe but globally, and how they have found in the AETE their home.

The preamble to Assisted Reproduction Technologies in Livestock

Many of the ARTs in use today were developed in Europe and disseminated around the world through exchange visit of scientists or veterinarians or during conferences. One very well know was the meeting organized by Tim Rowson at the animal research station in Cambridge, UK, in 1972, on the collection and surgical transfer of cattle embryos (Betteridge, 2003Betteridge KJ. A history of farm animal embryo transfer and some associated techniques. Anim Reprod Sci. 2003;79(3-4):203-44. http://doi.org/10.1016/S0378-4320(03)00166-0 PMid:14643106.
http://doi.org/10.1016/S0378-4320(03)001...
). Many of the attendees at this meeting were then the founders of IETS in 1974 in Colorado, where George Seidel also started a strong program at Colorado State University for the collection and transfer of cattle embryos. For about a decade, the focus was on the refinement of superovulatory protocols, which are substantially the same as those used today (Lonergan and Sánchez, 2022Lonergan P, Sánchez JM. 2022. Gamete and embryo technology: multiple ovulation and embryo transfer. In: McSweeney, P.L.H., McNamara, J.P., editors. Encyclopedia of dairy sciences. 3rd ed. Oxford: Academic Press, p. 881-889. http://doi.org/10.1016/B978-0-12-818766-1.00040-4.
http://doi.org/10.1016/B978-0-12-818766-...
), the optimization of the flushing and recovery protocol, and the replacement of the surgical embryo transfer with the non-surgical transcervical method (Wright, 1981Wright JM. Non-surgical embryo transfer in cattle embryo-recipient interactions. Theriogenology. 1981;15(1):43-56. http://doi.org/10.1016/S0093-691X(81)80017-9 PMid:16725539.
http://doi.org/10.1016/S0093-691X(81)800...
). The first successful cattle embryo cryopreservation was also achieved in Cambridge by Ian Wilmut (Wilmut and Rowson, 1973Wilmut I, Rowson LEA. Experiments on the low-temperature preservation of cow embryos. Vet Rec. 1973;92(26):686-90. http://doi.org/10.1136/vr.92.26.686 PMid:4730118.
http://doi.org/10.1136/vr.92.26.686...
) resulting in the birth of a calf named Frosty II.

Although the practitioners in the decade 1984-1994 were working to improve MOET (Multiple Ovulation and Embryo Transfer) or, more generally, in vivo derived embryos, many research laboratories concentrated on the in vitro production of embryos. Only a few years earlier, in1978, Louise Brown was born following in vitro fertilization performed by Robert Edwards (Steptoe and Edwards, 1978Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978;312(8085):366. http://doi.org/10.1016/S0140-6736(78)92957-4 PMid:79723.
http://doi.org/10.1016/S0140-6736(78)929...
), who conducted his experimental work at the Animal Research Station in Cambridge, again working with bovine oocytes. All these successful events in Europe served as a strong starting point for scientist and practitioners to continue their promising work.

Multiple ovulation and embryo transfer

The basis for the successful production of embryos in vivo is superovulation, and the understanding of the dynamic of follicular development is necessary to exploit the ovarian reserve of oocytes (Monniaux et al., 1983Monniaux D, Chupin D, Saumande J. Superovulatory responses of cattle. Theriogenology. 1983;19(1):55-81. http://doi.org/10.1016/0093-691X(83)90124-3.
http://doi.org/10.1016/0093-691X(83)9012...
, 2014Monniaux D, Clement F, Dalbies-Tran R, Estienne A, Fabre S, Mansanet C, Monget P. The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link? Biol Reprod. 2014;90(4):85. http://doi.org/10.1095/biolreprod.113.117077 PMid:24599291.
http://doi.org/10.1095/biolreprod.113.11...
; Monniaux, 2012Monniaux D. Superovulatory responses and embryo production in ruminants: lessons from ovary. In: 28th Annual Meeting A.E.T.E.; 2012 Sept 7-8; Saint Malo, France. Saint Malo: A.E.T.E.; 2012. p. 7-40.). Much of the work was done at INRA and often presented at AETE conferences. The protocol in the early days relied on the use of PMSG (now called eCG) but it had undesired side effects due to its long half -life (Monniaux et al., 1983Monniaux D, Chupin D, Saumande J. Superovulatory responses of cattle. Theriogenology. 1983;19(1):55-81. http://doi.org/10.1016/0093-691X(83)90124-3.
http://doi.org/10.1016/0093-691X(83)9012...
; Vos et al., 1994Vos PL, van der Schans A, de Wit AA, Bevers MM, Willemse AH, Dieleman SJ. Effects of neutralization of pregnant mares’ serum gonadotrophin (PMSG) shortly before or at the preovulatory LH surge in PMSG-superovulated heifers on follicular function and development. Reproduction. 1994;100(2):387-93. http://doi.org/10.1530/jrf.0.1000387 PMid:8021854.
http://doi.org/10.1530/jrf.0.1000387...
). A better understanding of follicular dynamics and follicular wave synchronization has allowed the optimization and development of more user-friendly protocols, but major advances in number of viable embryos produced have not been achieved (Bo and Mapletoft, 2014Bo GA, Mapletoft RJ. Historical perspectives and recent research on superovulation in cattle. Theriogenology. 2014;81(1):38-48. http://doi.org/10.1016/j.theriogenology.2013.09.020 PMid:24274408.
http://doi.org/10.1016/j.theriogenology....
). The development and use of recombinant b-FSH did not improve the results over the products extracted from pituitary glands (Wilson et al., 1993Wilson JM, Jones AL, Moore K, Looney CR, Bondioli KR. Superovulation of cattle with a recombinant-DNA bovine follicle stimulating hormone. Anim Reprod Sci. 1993;33(1-4):71-82. http://doi.org/10.1016/0378-4320(93)90107-3.
http://doi.org/10.1016/0378-4320(93)9010...
). Therefore, practitioners still rely today on pituitary extract of porcine origin (Folltropin, Pluset, Stimufol, 2 of these produced in Europe) or sheep origin (Ovagen) with the limitation that being a purified extract, there is inevitable batch to batch variation affecting their efficacy.

In vitro embryo production

The birth of the first baby by IVF sparked an interest in animal IVF, especially in livestock species, particularly in cattle. Although the first calf obtained by IVF was born in US using in vivo matured oocytes (Brackett et al., 1982Brackett BG, Bousquet D, Boice ML, Donawick WJ, Evans JF, Dressel MA. Normal development following in vitro fertilization in the cow. Biol Reprod. 1982;27(1):147-58. http://doi.org/10.1095/biolreprod27.1.147 PMid:6896830.
http://doi.org/10.1095/biolreprod27.1.14...
), the practical application required the use of immature oocytes harvested from ovaries and matured in vitro to metaphase II. Several European scientists contributed significantly to the in vitro maturation of livestock oocytes (Fulka et al., 1982Fulka J Jr, Pavlok A, Fulka J. In-vitro fertilization of zona-free bovine oocytes matured in culture. Reproduction. 1982;64(2):495-9. http://doi.org/10.1530/jrf.0.0640495 PMid:6896072.
http://doi.org/10.1530/jrf.0.0640495...
) demonstrating oocyte developmental competence (Staigmiller and Moor, 1984Staigmiller R, Moor R. Effect of follicle cells on the maturation and developmental competence of ovine oocytes matured outside the follicle. Gamete Res. 1984;9(2):221-9. http://doi.org/10.1002/mrd.1120090211.
http://doi.org/10.1002/mrd.1120090211...
) and finally, the culture of viable embryos in vitro (Gandolfi and Moor, 1987Gandolfi F, Moor RM. Stimulation of early embryonic development in the sheep by co-culture with oviduct epithelial cells. Reproduction. 1987;81(1):23-8. http://doi.org/10.1530/jrf.0.0810023 PMid:3668954.
http://doi.org/10.1530/jrf.0.0810023...
).

The potential value of in vitro technology quickly caught the interest of investors and the industry. Operations like Ovamass, associated with University College Dublin in Ireland, and Animal Biotechnology Cambridge on the Huntington Road premises in Cambridge, were established with the aim of producing large number of embryos from beef donors to be implanted into dairy cows. Similar operations were established in the Netherlands, France and Italy. Europe quickly became the leader in the production of embryos from slaughtered animals (Galli and Lazzari, 1996Galli C, Lazzari G. Practical aspects of IVM/IVF n cattle. Anim Reprod Sci. 1996;42(1-4):371-9. http://doi.org/10.1016/0378-4320(96)01530-8.
http://doi.org/10.1016/0378-4320(96)0153...
), as witnessed by the data published annually by the IETS Data Retrieval Committee.

The use of ovaries from slaughtered animals was very useful for research and for beef animals. However, from a genetic selection perspective, especially for dairy, it had to be done on live animals. In fact, in the years following the steps performed in the human field to obtain the first IVF baby also veterinarians started to practice ovum pick up on cows. The first attempts to use ultrasound guided follicular aspiration for embryo production in vitro were reported by Callesen et al. (1987) andCallesen H, Greve T, Christensen F. Ultrasonically guided aspiration of bovine follicular oocytes. Theriogenology. 1987;27(1):217. http://doi.org/10.1016/0093-691X(87)90094-X.
http://doi.org/10.1016/0093-691X(87)9009...
further developed by Pieterse et al. (1988Pieterse MC, Kappen KA, Kruip TAM, Taverne MAM. Aspiration of bovine oocytes during transvaginal ultrasound scanning of the ovaries. Theriogenology. 1988;30(4):751-62. http://doi.org/10.1016/0093-691X(88)90310-X PMid:16726517.
http://doi.org/10.1016/0093-691X(88)9031...
, 1991Pieterse MC, Vos PLAM, Kruip TAM, Wurth YA, van Beneden TH, Willemse AH, Taverne MAM. Transvaginal ultrasound guided follicular aspiration of bovine oocytes. Theriogenology. 1991;35(4):857-62. http://doi.org/10.1016/0093-691X(91)90426-E PMid:16726954.
http://doi.org/10.1016/0093-691X(91)9042...
). Using a human endovaginal probe adapted for the use in cattle, Pieterse reported a recovery rate of 55%, the repeatability of the procedure and the absence of side effects on the donor cows.

Although the procedures for embryo production in those days still required major laboratory refinements that came later on (Galli and Lazzari, 1996Galli C, Lazzari G. Practical aspects of IVM/IVF n cattle. Anim Reprod Sci. 1996;42(1-4):371-9. http://doi.org/10.1016/0378-4320(96)01530-8.
http://doi.org/10.1016/0378-4320(96)0153...
), the basics of OPU described by Pieterse et al. are still the same as those used today by many practitioners. Recovery rates have improved to over 70% due to the use of better ultrasound equipment with 6 or 7 MHz convex array probes that provide a better resolution on smaller follicles or the use of gonadotrophin priming that increases the size of smaller follicles. The OPU technique was initially applied on problem cows that did not respond to superovulation (Kruip et al., 1994Kruip TAM, Boni R, Wurth YA, Roelofsen MWM, Pieterse MC. Potential use of ovum pick-up for embryo production and breeding in cattle. Theriogenology. 1994;42(4):675-84. http://doi.org/10.1016/0093-691X(94)90384-U PMid:16727573.
http://doi.org/10.1016/0093-691X(94)9038...
; Looney et al., 1994Looney CR, Lindsey BR, Gonseth CL, Johnson DL. Commercial aspects of oocyte retrieval and in vitro fertilization (IVF) for embryo production in problem cows. Theriogenology. 1994;41(1):67-72. http://doi.org/10.1016/S0093-691X(05)80050-0.
http://doi.org/10.1016/S0093-691X(05)800...
), but it was later applied on a wider scale, including on pregnant cows, heifers and prepuberal heifers. (Galli et al., 2001Galli C, Crotti G, Notari C, Turini P, Duchi R, Lazzari G. Embryo production by ovum pick up from live donors. Theriogenology. 2001;55(6):1341-57. http://doi.org/10.1016/S0093-691X(01)00486-1 PMid:11327688.
http://doi.org/10.1016/S0093-691X(01)004...
). It is difficult and often not relevant to make comparisons between different data set since there are so many variables involved, most of which are not even manageable. Beef breeds perform better than dairy, dry cows do better than lactating ones and cows perform better than heifers. In vitro produced embryos cultured in presence of serum and/or co-culture had a reduced cryotolerance. For several years, until the culture media were improved, the surrogate sheep oviduct was used to produce freezable embryos (Rizos et al., 2002Rizos D, Ward F, Duffy P, Boland MP, Lonergan P. Consequences of bovine oocyte maturation, fertilization or early embryo development in vitro versus in vivo: implications for blastocyst yield and blastocyst quality. Mol Reprod Dev. 2002;61(2):234-48. http://doi.org/10.1002/mrd.1153 PMid:11803560.
http://doi.org/10.1002/mrd.1153...
; Lazzari et al., 2010Lazzari G, Colleoni S, Lagutina I, Crotti G, Turini P, Tessaro I, Brunetti D, Duchi R, Galli C. Short-term and long-term effects of embryo culture in the surrogate sheep oviduct versus in vitro culture for different domestic species. Theriogenology. 2010;73(6):748-57. http://doi.org/10.1016/j.theriogenology.2009.08.001 PMid:19726075.
http://doi.org/10.1016/j.theriogenology....
).

Associated with suboptimal in vitro culture systems the embryo developing in vitro were responsible for the so-called LOS (large offspring Syndrome), especially when embryos were originating from nuclear transfer and other invasive micromanipulations (Farin et al., 2010Farin CE, Farmer WT, Farin PW. Pregnancy recognition and abnormal offspring syndrome in cattle. Reprod Fertil Dev. 2010;22(1):75-87. http://doi.org/10.1071/RD09217 PMid:20003848.
http://doi.org/10.1071/RD09217...
). The underlying mechanisms were initially described by Young working in Edinburgh (Young et al., 1998Young LE, Sinclair KD, Wilmut I. Large offspring syndrome in cattle and sheep. Rev Reprod. 1998;3(3):155-63. http://doi.org/10.1530/ror.0.0030155 PMid:9829550.
http://doi.org/10.1530/ror.0.0030155...
; Lazzari et al., 2002bLazzari G, Wrenzycki C, Herrmann D, Duchi R, Kruip T, Niemann H, Galli C. Cellular and molecular deviations in bovine in vitro-produced embryos are related to the large offspring syndrome. Biol Reprod. 2002b;67(3):767-75. http://doi.org/10.1095/biolreprod.102.004481 PMid:12193383.
http://doi.org/10.1095/biolreprod.102.00...
). Due to the deregulation of imprinted genes, LOS resulted in offspring that was well above average birth weight, including placenta hypertrophy and hydroallantoids causing dystocia at parturition and increased stillbirth rate. Although the incidence of the phenomenon has decreased due to the better culture media devoid of fetal calf serum, it has not completely disappeared.

Although Europe led in the development of the technology and its practical application, in several AI organization and amongst practitioners, it did not follow the global trend whereby in vitro produced embryos are rapidly replacing in vivo derived ones today according to IETS data retrieval committee. In Europe, two-thirds of the bovine embryos produced still come from MOET, and in vitro produced embryos are mainly used by bull testing organizations.

Another species where in vitro embryo production is impacting breeding programs and practitioner activities is the horse. The implementation of ICSI (IntraCytoplasmic Sperm Injection), another technique developed in the human field (Palermo et al., 1992Palermo G, Joris H, Devroey P, Van Steirteghem AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet. 1992;340(8810):17-8. http://doi.org/10.1016/0140-6736(92)92425-F PMid:1351601.
http://doi.org/10.1016/0140-6736(92)9242...
) to bypass male infertility, found application in the horse to bypass zona hardening of the oocytes matured in vitro collected from the slaughterhouse or by ovum pick up (Lazzari et al., 2002aLazzari G, Crotti G, Turini P, Duchi R, Mari G, Zavaglia G, Barbacini S, Galli C. Equine embryos at the compacted morula and blastocyst stage can be obtained by intracytoplasmic sperm injection (ICSI) of in vitro matured oocytes with frozen-thawed spermatozoa from semen of different fertilities. Theriogenology. 2002a;58(2-4):709-12. http://doi.org/10.1016/S0093-691X(02)00777-X.
http://doi.org/10.1016/S0093-691X(02)007...
; Galli et al., 2007Galli C, Colleoni S, Duchi R, Lagutina I, Lazzari G. Developmental competence of equine oocytes and embryos obtained by in vitro procedures ranging from in vitro maturation and ICSI to embryo culture, cryopreservation and somatic cell nuclear transfer. Anim Reprod Sci. 2007;98(1-2):39-55. http://doi.org/10.1016/j.anireprosci.2006.10.011 PMid:17101246.
http://doi.org/10.1016/j.anireprosci.200...
). ICSI is revolutionizing the horse breeding industry because of difficulties in capacitating the stallion spermatozoa, the low quality of many frozen semen samples or the limited availability of semen of dead stallions in addition to the sub-fertile or old mares unusable by conventional in vivo flushing (Lazzari et al., 2020Lazzari G, Colleoni S, Crotti G, Turini P, Fiorini G, Barandalla M, Landriscina L, Dolci G, Benedetti M, Duchi R, Galli C. Laboratory production of equine embryos. J Equine Vet Sci. 2020;89:103097. http://doi.org/10.1016/j.jevs.2020.103097 PMid:32563445.
http://doi.org/10.1016/j.jevs.2020.10309...
; Claes and Stout, 2022Claes A, Stout TAE. Success rate in a clinical equine in vitro embryo production program. Theriogenology. 2022;187:215-8. http://doi.org/10.1016/j.theriogenology.2022.04.019 PMid:35623226.
http://doi.org/10.1016/j.theriogenology....
), where once again Europe is leading the way. According to the AETE Data Retrieval Committee, the number of in vitro produced horse embryos is greater than the one produced by in vivo flushing. This is also supported by the competitive advantage that ICSI embryos can be cryopreserved very successfully, both by slow freezing or vitrification, making it possible for a seasonal breeder like the horse to produce embryos also outside the breeding season for transfer during the breeding season or for marketing. The pregnancies and the foals obtained from ICSI embryos are normal and do not exhibit phenotypical abnormalities like the LOS observed in ruminants. This technique is also rapidly developing both in South and North America, as it has for bovines.

Embryo/semen sexing and Genomic selection of livestock

Having the offspring of the desired sex has always been the desire of all breeders. When PCR came on the market in 1988, sexing of cattle embryos became a reality for many cattle breeding organization, especially in Europe for dairy breeds (Bredbacka et al., 1995Bredbacka P, Kankaanpaa A, Peippo J. PCR-sexing of bovine embryos: a simplified protocol. Theriogenology. 1995;44(2):167-76. http://doi.org/10.1016/0093-691X(95)00166-6 PMid:16727716.
http://doi.org/10.1016/0093-691X(95)0016...
; Thibier and Nibart, 1995Thibier M, Nibart M. The sexing of bovine embryos in the field. Theriogenology. 1995;43(1):71-80. http://doi.org/10.1016/0093-691X(94)00008-I.
http://doi.org/10.1016/0093-691X(94)0000...
), and also for individual practitioners when portable kits and simplified protocols were developed for field use. The procedure required to take a biopsy from the embryos (5 to 10 cells) had to be done carefully w/o damaging the embryo too much, and the embryos and fresh transfer was the preferred protocol. Embryo sexing is also used in horses where there is no sexed semen available at the commercial level (Lazzari et al., 2020Lazzari G, Colleoni S, Crotti G, Turini P, Fiorini G, Barandalla M, Landriscina L, Dolci G, Benedetti M, Duchi R, Galli C. Laboratory production of equine embryos. J Equine Vet Sci. 2020;89:103097. http://doi.org/10.1016/j.jevs.2020.103097 PMid:32563445.
http://doi.org/10.1016/j.jevs.2020.10309...
; Coster et al., 2023Coster T, Van Poucke M, Bogado Pascottini O, Angel-Velez D, Van den Branden E, Peere S, Papas M, Gerits I, Govaere J, Peelman L, Vermeesch JR, Van Soom A, Smits K. Single closed-tube quantitative real-time PCR assay with dual-labelled probes for improved sex determination of equine embryos. Animal. 2023;17(11):100952. http://doi.org/10.1016/j.animal.2023.100952 PMid:37913607.
http://doi.org/10.1016/j.animal.2023.100...
). While embryo sexing allows to know the sex of the embryo, the use of sexed semen predetermines the sex of the embryo allowing the production of the desired sex only. The refinements and the commercialization of the semen sexing technology was later done in US but the initial groundbreaking experiments on separating X and Y sperm were performed by Jane Morrell (current Board Member of AETE) at the National Institute for Medical Research in London (Morrell et al., 1988Morrell JM, Keeler KD, Noakes DE, Mackenzie NM, Dresser DW. Sexing of sperm by flow cytometry. Vet Rec. 1988;122(14):322-4. http://doi.org/10.1136/vr.122.14.322 PMid:3164152.
http://doi.org/10.1136/vr.122.14.322...
). The technique of embryo biopsy, superseded by sexed semen for sex selection, has remained relevant with the introduction of genomic selection (Hayes et al., 2009Hayes BJ, Bowman PJ, Chamberlain AJ, Goddard ME. Invited review: Genomic selection in dairy cattle: progress and challenges. J Dairy Sci. 2009;92(2):433-43. http://doi.org/10.3168/jds.2008-1646 PMid:19164653.
http://doi.org/10.3168/jds.2008-1646...
). Several cattle breeding organization in Europe were quick to implement genomic selection of embryos before transfer or freezing (Ponsart et al., 2014Ponsart C, Le Bourhis D, Knijn H, Fritz S, Guyader-Joly C, Otter T, Lacaze S, Charreaux F, Schibler L, Dupassieux D, Mullaart E. Reproductive technologies and genomic selection in dairy cattle. Reprod Fertil Dev. 2014;26(1):12-21. http://doi.org/10.1071/RD13328 PMid:24305173.
http://doi.org/10.1071/RD13328...
) to accelerate selection primarily on the male line, to select the bull of the next generation and avoid the birth of unwanted bull calves. Similar work has been undertaken also on equine embryos (Coster et al., 2024Coster T, Zhao Y, Tsuiko O, Demyda-Peyras S, Van Soom A, Vermeesch JR, Smits K. Genome-wide equine preimplantation genetic testing enabled by simultaneous haplotyping and copy number detection. Sci Rep. 2024;14(1):2003. http://doi.org/10.1038/s41598-023-48103-7 PMid:38263320.
http://doi.org/10.1038/s41598-023-48103-...
).

Cloning by nuclear transfer

Another dream of the animal breeders was to achieve the quality and uniformity typical of plant breeders where cloning is widely used. Cloning mammals is more complicated and the first experiments were actually performed in amphibians (Gurdon, 1962Gurdon JB. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. Development. 1962;10(4):622-40. http://doi.org/10.1242/dev.10.4.622 PMid:13951335.
http://doi.org/10.1242/dev.10.4.622...
). Despite attempts by many laboratories to clone mice and a controversial publication by an Austrian investigators (Illmensee and Hoppe, 1981Illmensee K, Hoppe PC. Nuclear transplantation in Mus musculus: developmental potential of nuclei from preimplantation embryos. Cell. 1981;23(1):9-18. http://doi.org/10.1016/0092-8674(81)90265-8 PMid:7214529.
http://doi.org/10.1016/0092-8674(81)9026...
) claiming success, the mouse turned out to be more difficult to clone than livestock.

Steen Willadsen a Danish veterinarian working in Cambridge (an AETE pioneer awardee), was a key player in cloning sheep and cattle both by blastomere separation and by nuclear transfer (Willadsen, 1986Willadsen SM. Nuclear transplantation in sheep embryos. Nature. 1986;320(6057):63-5. http://doi.org/10.1038/320063a0 PMid:3951549.
http://doi.org/10.1038/320063a0...
). Embryo cloning, as developed by Willadsen, had clear limitations on the number of nuclei available in each morula (20 to 30 cells) used, and the process of serial cloning had limitation after the first round. Despite this, embryo cloning was taken up by newly established cloning companies interested in cattle breeding in North America. However, it became clear that, together with the technical difficulties, the phenotype of the embryo was unpredictable, and the interest waned.

It was again thanks to European scientists, with the cloning of Dolly the sheep (Wilmut et al., 1997Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997;385(6619):810-3. http://doi.org/10.1038/385810a0 PMid:9039911.
http://doi.org/10.1038/385810a0...
) that cloning, or better defined as Somatic Cell Nuclear Transfer, regained attention. The possibility to clone an adult animal of known phenotype clearly makes the difference and re-ignited the interest of the industry as well as scientists. After Dolly several other mammals were cloned from somatic cells in Europe including the bovine (Galli et al., 1999Galli C, Duchi R, Moor RM, Lazzari G. Mammalian leukocytes contain all the genetic information necessary for the development of a new individual. Cloning. 1999;1(3):161-70. http://doi.org/10.1089/15204559950019924 PMid:16218815.
http://doi.org/10.1089/15204559950019924...
), the horse (Galli et al., 2003Galli C, Lagutina I, Crotti G, Colleoni S, Turini P, Ponderato N, Duchi R, Lazzari G. Pregnancy: a cloned horse born to its dam twin. Nature. 2003;424(6949):635. http://doi.org/10.1038/424635a PMid:12904778.
http://doi.org/10.1038/424635a...
), the rat (Zhou et al., 2003Zhou Q, Renard JP, Le Friec G, Brochard V, Beaujean N, Cherifi Y, Fraichard A, Cozzi J. Generation of fertile cloned rats by regulating oocyte activation. Science. 2003;302(5648):1179. http://doi.org/10.1126/science.1088313 PMid:14512506.
http://doi.org/10.1126/science.1088313...
), the mouflon through interspecies nuclear transfer (Loi et al., 2001Loi P, Ptak G, Barboni B, Fulka J Jr, Cappai P, Clinton M. Genetic rescue of an endangered mammal by cross-species nuclear transfer using post-mortem somatic cells. Nat Biotechnol. 2001;19(10):962-4. http://doi.org/10.1038/nbt1001-962 PMid:11581663.
http://doi.org/10.1038/nbt1001-962...
) to mention a few.

Studies were also undertaken, especially by Yvan Heyman (Heyman et al., 2007Heyman Y, Chavatte-Palmer P, Berthelot V, Fromentin G, Hocquette JF, Martignat L, Renard JP. Assessing the quality of products from cloned cattle: an integrative approach. Theriogenology. 2007;67(1):134-41. http://doi.org/10.1016/j.theriogenology.2006.09.020 PMid:17092550.
http://doi.org/10.1016/j.theriogenology....
) to demonstrate that the products originating from cloned animals did not differ from non-cloned controls. Despite all these efforts and the pioneering role of many European scientists, cloned animals and their products are not allowed to enter the food chain in Europe. Cloning by somatic cell nuclear transfer is still not efficient, especially in ruminants but works better in pigs and horses.

The reprogramming of the genome of a differentiated cells provided an unprecedented opportunity for scientists interested in understanding the epigenetic events underlying cell differentiation (Yang et al., 2007Yang X, Smith SL, Tian XC, Lewin HA, Renard JP, Wakayama T. Nuclear reprogramming of cloned embryos and its implications for therapeutic cloning. Nat Genet. 2007;39(3):295-302. http://doi.org/10.1038/ng1973 PMid:17325680.
http://doi.org/10.1038/ng1973...
; Matoba and Zhang, 2018Matoba S, Zhang Y. Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell. 2018;23(4):471-85. http://doi.org/10.1016/j.stem.2018.06.018 PMid:30033121.
http://doi.org/10.1016/j.stem.2018.06.01...
). The unravelling of the mechanisms involved in genome differentiation and reprogramming will be important to increase the efficiency of SCNT. However, these advancements will probably not come from European scientists or industry since the funding of the EU that supported most of the past European successes described above is no more available and directed to other “politically correct” priorities.

Stem cells and genetic engineering

In 1981 Martin Evans, working at Cambridge, UK, published a seminal paper to describe the derivation of embryonic stem (ES) cells from the mouse embryo (Evans and Kaufman, 1981Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292(5819):154-6. http://doi.org/10.1038/292154a0 PMid:7242681.
http://doi.org/10.1038/292154a0...
). This work earned him the Nobel prize in 2007 shared with Mario Capecchi and Oliver Smithies for the development of “gene targeting”, concept largely used today for genome editing. The use of embryonic stem cells became fundamental to generate the knock out mouse models to understand the function of any given gene in the genome (Robertson et al., 1986Robertson E, Bradley A, Kuehn M, Evans M. Germ-line transmission of genes introduced into cultured pluripotential cells by retroviral vector. Nature. 1986;323(6087):445-8. http://doi.org/10.1038/323445a0 PMid:3762693.
http://doi.org/10.1038/323445a0...
).

Given the potential of embryonic stem cells several laboratories attempted to derive ES cells from livestock species (Notarianni et al., 1991Notarianni E, Galli C, Laurie S, Moor RM, Evans MJ. Derivation of pluripotent, embryonic cell lines from the pig and sheep. J Reprod Fertil Suppl. 1991;43:255-60. PMid:1843344.) as it would provide an unlimited source of cells for cloning. However, it turned out to be a daunting task (Galli et al., 1994Galli C, Lazzari G, Flechon JE, Moor RM. Embryonic stem cells in farm animals. Zygote. 1994;2(4):385-9. http://doi.org/10.1017/S0967199400002288 PMid:8665176.
http://doi.org/10.1017/S0967199400002288...
). Interest in stem cells was also driven by the possibility of genetic engineering as it was done in the mouse, but the molecular pathways were only partially understood (Lazzari et al., 2006Lazzari G, Colleoni S, Giannelli SG, Brunetti D, Colombo E, Lagutina I, Galli C, Broccoli V. Direct derivation of neural rosettes from cloned bovine blastocysts: a model of early neurulation events and neural crest specification in vitro. Stem Cells. 2006;24(11):2514-21. http://doi.org/10.1634/stemcells.2006-0149 PMid:16931774.
http://doi.org/10.1634/stemcells.2006-01...
). The undifferentiated state could only be kept for a limited time in culture and it was not until the conditions for human ES cells were worked out (Thomson et al., 1998Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145-7. http://doi.org/10.1126/science.282.5391.1145 PMid:9804556.
http://doi.org/10.1126/science.282.5391....
) that the derivation of stable bovine ES cells was reported (Bogliotti et al., 2018Bogliotti YS, Wu J, Vilarino M, Okamura D, Soto DA, Zhong C, Sakurai M, Sampaio RV, Suzuki K, Izpisua Belmonte JC, Ross PJ. Efficient derivation of stable primed pluripotent embryonic stem cells from bovine blastocysts. Proc Natl Acad Sci USA. 2018;115(9):2090-5. http://doi.org/10.1073/pnas.1716161115 PMid:29440377.
http://doi.org/10.1073/pnas.1716161115...
). Currently the interests in livestock ES cells is mainly academic since cloning can be done with somatic cells and it appears that there is no advantage to using less differentiated cells for nuclear transfer compared to fully differentiated ones (Sung et al., 2006Sung LY, Gao S, Shen H, Yu H, Song Y, Smith SL, Chang CC, Inoue K, Kuo L, Lian J, Li A, Tian XC, Tuck DP, Weissman SM, Yang X, Cheng T. Differentiated cells are more efficient than adult stem cells for cloning by somatic cell nuclear transfer. Nat Genet. 2006;38(11):1323-8. http://doi.org/10.1038/ng1895 PMid:17013394.
http://doi.org/10.1038/ng1895...
).

The interest in generating livestock carrying genetic modification was present also in Europe after the pioneering work in USA of Brinster (Hammer et al., 1985Hammer RE, Pursel VG, Rexroad CE Jr, Wall RJ, Bolt DJ, Ebert KM, Palmiter RD, Brinster RL. Production of transgenic rabbits, sheep and pigs by microinjection. Nature. 1985;315(6021):680-3. http://doi.org/10.1038/315680a0 PMid:3892305.
http://doi.org/10.1038/315680a0...
) by microinjecting the pronucleus of the zygote as it was done in the mouse. Several animals carrying transgenes of pharmaceutical interest were generated (Clark et al., 1989Clark AJ, Bessos H, Bishop JO, Brown P, Harris S, Lathe R, McClenaghan M, Prowse C, Simons JP, Whitelaw CBA, Wilmut I. Expression of human anti-hemophilic factor IX in the milk of transgenic sheep. Bio/Technology. 1989;7:487-92.; Niemann et al., 1996Niemann H, Halter R, Espanion G, Wrenzycki C, Herrmann D, Lemme E, Carnwath JW 1, Paul D. Expression of human blood clotting factor VIII (FVIII) constructs in the mammary gland of transgenic mice and sheep. J Anim Breed Genet. 1996;113(1-6):437-44. http://doi.org/10.1111/j.1439-0388.1996.tb00634.x.
http://doi.org/10.1111/j.1439-0388.1996....
) but the efficiency of the system was low making the projects very expensive and, in the long term, unsustainable.

The breakthrough to generate genome edited animals came with the discovery of programmable nucleases in the last ten to fifteen years. First, the Zinc fingers nucleases (Urnov et al., 2010Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11(9):636-46. http://doi.org/10.1038/nrg2842 PMid:20717154.
http://doi.org/10.1038/nrg2842...
), then the TALENs (Joung and Sander, 2013Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2013;14(1):49-55. http://doi.org/10.1038/nrm3486 PMid:23169466.
http://doi.org/10.1038/nrm3486...
) and the Crispr/Cas9 (Jinek et al., 2012Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-21. http://doi.org/10.1126/science.1225829 PMid:22745249.
http://doi.org/10.1126/science.1225829...
) opened a new era in the genetic modification of animals and plants. Unfortunately, most of the work behind these developments and applications took place in North America as in Europe the “phobia” against GMOs has cut the funding to scientists, driven away companies and investors. Interestingly, the basic discovery behind the CRISPR/Cas9 technology was done by F.J. Mojica a Spanish scientist working in Alicante (Mojica et al., 1993Mojica FJ, Juez G, Rodriguez-Valera F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol Microbiol. 1993;9(3):613-21. http://doi.org/10.1111/j.1365-2958.1993.tb01721.x PMid:8412707.
http://doi.org/10.1111/j.1365-2958.1993....
; Lander, 2016Lander ES. The Heroes of CRISPR. Cell. 2016;164(1-2):18-28. http://doi.org/10.1016/j.cell.2015.12.041 PMid:26771483.
http://doi.org/10.1016/j.cell.2015.12.04...
).

On the 25th anniversary of the birth of Dolly (Galli and Lazzari, 2021Galli C, Lazzari G. 25th Anniversary of Cloning by Somatic-Cell Nuclear Transfer: current applications of SCNT in advanced breeding and genome editing in livestock. Reproduction. 2021;162(1):F23-f32. PMid:33852430.) we are witnessing a revision of the European policy on these techniques, defined as New Genomic Techniques (NGT) (https://ec.europa.eu/food/plant/gmo/modern_biotech/new-genomic-techniques_en). Someone would say better late than never. This revision is primarily considered for plants but still not for animals despite the vast number of livestock genome edited already generated (Bishop and Van Eenennaam, 2020Bishop TF, Van Eenennaam AL. Genome editing approaches to augment livestock breeding programs. J Exp Biol. 2020;223(Suppl. 1):jeb207159. http://doi.org/10.1242/jeb.207159 PMid:32034040.
http://doi.org/10.1242/jeb.207159...
) for agricultural purposes in many parts of the world except Europe.

Currently in Europe we are using genome editing techniques in the field of xenotransplantation to create pigs whose organs, tissues or cells could be transplanted to humans (Fischer and Schnieke, 2022Fischer K, Schnieke A. Xenotransplantation becoming reality. Transgenic Res. 2022;31(3):391-8. http://doi.org/10.1007/s11248-022-00306-w PMid:35545691.
http://doi.org/10.1007/s11248-022-00306-...
; Galli, 2023Galli C. Animal engineering for xenotransplantation. EJT. 2023;1(3):182-91. http://doi.org/10.57603/EJT-307.
http://doi.org/10.57603/EJT-307...
) or to generate animal models of human genetic diseases (Aigner et al., 2010Aigner B, Renner S, Kessler B, Klymiuk N, Kurome M, Wunsch A, Wolf E. Transgenic pigs as models for translational biomedical research. J Mol Med (Berl). 2010;88(7):653-64. http://doi.org/10.1007/s00109-010-0610-9 PMid:20339830.
http://doi.org/10.1007/s00109-010-0610-9...
; Porta-Sanchez et al., 2023Porta-Sanchez A, Mazzanti A, Tarifa C, Kukavica D, Trancuccio A, Mohsin M, Zanfrini E, Perota A, Duchi R, Hernandez-Lopez K, Jauregui-Abularach ME, Pergola V, Fernandez E, Bongianino R, Tavazzani E, Gambelli P, Memmi M, Scacchi S, Pavarino LF, Franzone PC, Lentini G, Filgueiras-Rama D, Galli C, Santiago DJ, Priori SG. Unexpected impairment of I(Na) underpins reentrant arrhythmias in a knock-in swine model of Timothy syndrome. Nat Cardiovasc Res. 2023;2(12):1291-309. http://doi.org/10.1038/s44161-023-00393-w PMid:38665938.
http://doi.org/10.1038/s44161-023-00393-...
). All this work is at the R&D phase, as the regulatory pathway for approval through the regulatory agencies has yet to be tested.

Assisted Reproduction Technologies for conservation biology

An area where ARTs are put at work at its best and Europe is leading the way is for the Biorescue project (https://www.biorescue.org/), a race against time to save the Northern White Rhino, an iconic species where only two female are living on earth and are based in Kenya. This project besides being unique in its scope it will also serve as a template for other endangered species. Several European institutions are involved in this project covering the clinical area on live animals for oocyte recovery and embryo transfer (Leibniz Institute for Zoo and Wildlife Research, Berlin), the politics and logistic (Safari Park Dvůr Králové, Czech Republic), embryo production (Avantea, Cremona), stem cell biology (Max Delbrück Center for Molecular Medicine) and ethic (Department of Comparative Biomedicine and Food Science, Padova). To date the project has been very successful with the initial trials with Southern White Rhino females to develop and validate the technique that produced embryos and two lines of ES cells (Hildebrandt et al., 2018Hildebrandt TB, Hermes R, Colleoni S, Diecke S, Holtze S, Renfree MB, Stejskal J, Hayashi K, Drukker M, Loi P, Goritz F, Lazzari G, Galli C. Embryos and embryonic stem cells from the white rhinoceros. Nat Commun. 2018;9(1):2589. http://doi.org/10.1038/s41467-018-04959-2 PMid:29973581.
http://doi.org/10.1038/s41467-018-04959-...
). In 2019 we initiated the OPU and embryo production on the two NWR female left (Najin and Fatu, mother, and daughter). Soon we realized that only the daughter was producing embryos therefore we stopped collecting the mother. To date we have produced and cryopreserved 30 NWR embryos and many more from SWR (Hildebrandt et al., 2023Hildebrandt TB, Holtze S, Colleoni S, Hermes R, Stejskal J, Lekolool I, Ndeereh D, Omondi P, Kariuki L, Mijele D, Mutisya S, Ngulu S, Diecke S, Hayashi K, Lazzari G, de Mori B, Biasetti P, Quaggio A, Galli C, Goeritz F. In vitro fertilization program in white rhinoceros. Reproduction. 2023;166(6):383-99. http://doi.org/10.1530/REP-23-0087 PMid:37877686.
http://doi.org/10.1530/REP-23-0087...
). In the meantime, we have been working to develop embryo transfer. The challenges are many including the preparation of a vasectomized teaser bull to detect exactly when the surrogate recipients are in estrus. To date one pregnancy has been established with a SWR embryo (unpublished). To widen the genetic base to be able to have a self-sustaining population we are also using stem cell technologies to generate oocytes and spermatozoa in vitro through iPSc (induced pluripotent stem cells) (Hayashi et al., 2022Hayashi M, Zywitza V, Naitou Y, Hamazaki N, Goeritz F, Hermes R, Holtze S, Lazzari G, Galli C, Stejskal J, Diecke S, Hildebrandt TB, Hayashi K. Robust induction of primordial germ cells of white rhinoceros on the brink of extinction. Sci Adv. 2022;8(49):eabp9683. http://doi.org/10.1126/sciadv.abp9683 PMid:36490332.
http://doi.org/10.1126/sciadv.abp9683...
; Zywitza et al., 2022Zywitza V, Rusha E, Shaposhnikov D, Ruiz-Orera J, Telugu N, Rishko V, Hayashi M, Michel G, Wittler L, Stejskal J, Holtze S, Göritz F, Hermes R, Wang J, Izsvák Z, Colleoni S, Lazzari G, Galli C, Hildebrandt TB, Hayashi K, Diecke S, Drukker M. Naïve-like pluripotency to pave the way for saving the northern white rhinoceros from extinction. Sci Rep. 2022;12(1):3100. http://doi.org/10.1038/s41598-022-07059-w PMid:35260583.
http://doi.org/10.1038/s41598-022-07059-...
). The application of ART in conservation biology is viewed with suspicion by the stakeholders who, for a long time, opposed their introduction. Therefore, it is imperative that an ethical assessment is in place before, during, and after the procedures are performed, both on the animals and in the laboratory (Mori et al., 2021Mori B, Spiriti MM, Pollastri I, Normando S, Biasetti P, Florio D, Andreucci F, Colleoni S, Galli C, Goritz F, Hermes R, Holtze S, Lazzari G, Seet S, Zwilling J, Stejskal J, Mutisya S, Ndeereh D, Ngulu S, Vigne R, Hildebrandt TB. An Ethical Assessment Tool (ETHAS) to evaluate the application of assisted reproductive technologies in mammals’ conservation: the case of the northern white rhinoceros (Ceratotherium simum cottoni). Animals (Basel). 2021;11(2):311. http://doi.org/10.3390/ani11020312 PMid:33530613.
http://doi.org/10.3390/ani11020312...
; Biasetti et al., 2022Biasetti P, Hildebrandt TB, Goritz F, Hermes R, Holtze S, Galli C, Lazzari G, Colleoni S, Pollastri I, Spiriti MM, Stejskal J, Seet S, Zwilling J, Ngulu S, Mutisya S, Kariuki L, Lokolool I, Omondo P, Ndeereh D, de Mori B. Ethical analysis of the application of assisted reproduction technologies in biodiversity conservation and the case of white rhinoceros (Ceratotherium simum) ovum pick-up procedures. Front Vet Sci. 2022;9:831675. http://doi.org/10.3389/fvets.2022.831675 PMid:35591869.
http://doi.org/10.3389/fvets.2022.831675...
).

Final considerations

ARTs and related techniques have made huge progress in the past 40 years, both in livestock and wildlife species. This progress has been fostered by several circumstances. First, by the public funding made available at the national and, above all, the European level. This has facilitated the propensity of laboratories to exchange scientists, collaborations between research groups, and presentations of original work at conferences. In the last decade or more, such funding for livestock research is no longer available, hastening competition rather than collaboration, as the driving forces to attract funding are now the number of publications or the number of patents at the expense of innovative, reproducible, and sharable work. Second, by the number of public institutions and practitioners, with companies or cooperatives that created a critical mass of knowledge and work with direct practical implication that required solutions. Third, the scientific societies like AETE with annual meetings fostered the exchange of ideas, discussions with regulators and interactions in presence between members that, with the digital era and the recent pandemic, had suffered a lot.

Looking ahead the prospects are not very optimistic. Alongside the reduction of funding, there is also a growing opposition to animal breeding. This opposition arises not only for ethical reasons but also due to concerns about environmental impact and other trendy topics in today’s political discussions. Unfortunately, these discussions often overlook the role of assisted reproduction in a broader context, including its significance in human fields, which are strongly interconnected. As for the future, I believe it will largely be in the hands of the younger generations. While they are being trained with modern techniques and tools, it's important for them not to forget the lessons of the past. By learning from history, they can better plan for the future using the new techniques and instruments available today.

  • Financial support: None.
  • How to cite: Galli C, Lazzari G. 40 years of AETE: the contribution of scientists and practitioners to the progress of reproductive biotechnologies in Europe. Anim Reprod. 2024;21(3):e20240061. https://doi.org/10.1590/1984-3143-AR2024-0061

References

  • Aigner B, Renner S, Kessler B, Klymiuk N, Kurome M, Wunsch A, Wolf E. Transgenic pigs as models for translational biomedical research. J Mol Med (Berl). 2010;88(7):653-64. http://doi.org/10.1007/s00109-010-0610-9 PMid:20339830.
    » http://doi.org/10.1007/s00109-010-0610-9
  • Betteridge KJ. A history of farm animal embryo transfer and some associated techniques. Anim Reprod Sci. 2003;79(3-4):203-44. http://doi.org/10.1016/S0378-4320(03)00166-0 PMid:14643106.
    » http://doi.org/10.1016/S0378-4320(03)00166-0
  • Biasetti P, Hildebrandt TB, Goritz F, Hermes R, Holtze S, Galli C, Lazzari G, Colleoni S, Pollastri I, Spiriti MM, Stejskal J, Seet S, Zwilling J, Ngulu S, Mutisya S, Kariuki L, Lokolool I, Omondo P, Ndeereh D, de Mori B. Ethical analysis of the application of assisted reproduction technologies in biodiversity conservation and the case of white rhinoceros (Ceratotherium simum) ovum pick-up procedures. Front Vet Sci. 2022;9:831675. http://doi.org/10.3389/fvets.2022.831675 PMid:35591869.
    » http://doi.org/10.3389/fvets.2022.831675
  • Bishop TF, Van Eenennaam AL. Genome editing approaches to augment livestock breeding programs. J Exp Biol. 2020;223(Suppl. 1):jeb207159. http://doi.org/10.1242/jeb.207159 PMid:32034040.
    » http://doi.org/10.1242/jeb.207159
  • Bo GA, Mapletoft RJ. Historical perspectives and recent research on superovulation in cattle. Theriogenology. 2014;81(1):38-48. http://doi.org/10.1016/j.theriogenology.2013.09.020 PMid:24274408.
    » http://doi.org/10.1016/j.theriogenology.2013.09.020
  • Bogliotti YS, Wu J, Vilarino M, Okamura D, Soto DA, Zhong C, Sakurai M, Sampaio RV, Suzuki K, Izpisua Belmonte JC, Ross PJ. Efficient derivation of stable primed pluripotent embryonic stem cells from bovine blastocysts. Proc Natl Acad Sci USA. 2018;115(9):2090-5. http://doi.org/10.1073/pnas.1716161115 PMid:29440377.
    » http://doi.org/10.1073/pnas.1716161115
  • Brackett BG, Bousquet D, Boice ML, Donawick WJ, Evans JF, Dressel MA. Normal development following in vitro fertilization in the cow. Biol Reprod. 1982;27(1):147-58. http://doi.org/10.1095/biolreprod27.1.147 PMid:6896830.
    » http://doi.org/10.1095/biolreprod27.1.147
  • Bredbacka P, Kankaanpaa A, Peippo J. PCR-sexing of bovine embryos: a simplified protocol. Theriogenology. 1995;44(2):167-76. http://doi.org/10.1016/0093-691X(95)00166-6 PMid:16727716.
    » http://doi.org/10.1016/0093-691X(95)00166-6
  • Callesen H, Greve T, Christensen F. Ultrasonically guided aspiration of bovine follicular oocytes. Theriogenology. 1987;27(1):217. http://doi.org/10.1016/0093-691X(87)90094-X
    » http://doi.org/10.1016/0093-691X(87)90094-X
  • Claes A, Stout TAE. Success rate in a clinical equine in vitro embryo production program. Theriogenology. 2022;187:215-8. http://doi.org/10.1016/j.theriogenology.2022.04.019 PMid:35623226.
    » http://doi.org/10.1016/j.theriogenology.2022.04.019
  • Clark AJ, Bessos H, Bishop JO, Brown P, Harris S, Lathe R, McClenaghan M, Prowse C, Simons JP, Whitelaw CBA, Wilmut I. Expression of human anti-hemophilic factor IX in the milk of transgenic sheep. Bio/Technology. 1989;7:487-92.
  • Coster T, Van Poucke M, Bogado Pascottini O, Angel-Velez D, Van den Branden E, Peere S, Papas M, Gerits I, Govaere J, Peelman L, Vermeesch JR, Van Soom A, Smits K. Single closed-tube quantitative real-time PCR assay with dual-labelled probes for improved sex determination of equine embryos. Animal. 2023;17(11):100952. http://doi.org/10.1016/j.animal.2023.100952 PMid:37913607.
    » http://doi.org/10.1016/j.animal.2023.100952
  • Coster T, Zhao Y, Tsuiko O, Demyda-Peyras S, Van Soom A, Vermeesch JR, Smits K. Genome-wide equine preimplantation genetic testing enabled by simultaneous haplotyping and copy number detection. Sci Rep. 2024;14(1):2003. http://doi.org/10.1038/s41598-023-48103-7 PMid:38263320.
    » http://doi.org/10.1038/s41598-023-48103-7
  • Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292(5819):154-6. http://doi.org/10.1038/292154a0 PMid:7242681.
    » http://doi.org/10.1038/292154a0
  • Farin CE, Farmer WT, Farin PW. Pregnancy recognition and abnormal offspring syndrome in cattle. Reprod Fertil Dev. 2010;22(1):75-87. http://doi.org/10.1071/RD09217 PMid:20003848.
    » http://doi.org/10.1071/RD09217
  • Fischer K, Schnieke A. Xenotransplantation becoming reality. Transgenic Res. 2022;31(3):391-8. http://doi.org/10.1007/s11248-022-00306-w PMid:35545691.
    » http://doi.org/10.1007/s11248-022-00306-w
  • Fulka J Jr, Pavlok A, Fulka J. In-vitro fertilization of zona-free bovine oocytes matured in culture. Reproduction. 1982;64(2):495-9. http://doi.org/10.1530/jrf.0.0640495 PMid:6896072.
    » http://doi.org/10.1530/jrf.0.0640495
  • Galli C, Colleoni S, Duchi R, Lagutina I, Lazzari G. Developmental competence of equine oocytes and embryos obtained by in vitro procedures ranging from in vitro maturation and ICSI to embryo culture, cryopreservation and somatic cell nuclear transfer. Anim Reprod Sci. 2007;98(1-2):39-55. http://doi.org/10.1016/j.anireprosci.2006.10.011 PMid:17101246.
    » http://doi.org/10.1016/j.anireprosci.2006.10.011
  • Galli C, Crotti G, Notari C, Turini P, Duchi R, Lazzari G. Embryo production by ovum pick up from live donors. Theriogenology. 2001;55(6):1341-57. http://doi.org/10.1016/S0093-691X(01)00486-1 PMid:11327688.
    » http://doi.org/10.1016/S0093-691X(01)00486-1
  • Galli C, Duchi R, Moor RM, Lazzari G. Mammalian leukocytes contain all the genetic information necessary for the development of a new individual. Cloning. 1999;1(3):161-70. http://doi.org/10.1089/15204559950019924 PMid:16218815.
    » http://doi.org/10.1089/15204559950019924
  • Galli C, Lagutina I, Crotti G, Colleoni S, Turini P, Ponderato N, Duchi R, Lazzari G. Pregnancy: a cloned horse born to its dam twin. Nature. 2003;424(6949):635. http://doi.org/10.1038/424635a PMid:12904778.
    » http://doi.org/10.1038/424635a
  • Galli C, Lazzari G, Flechon JE, Moor RM. Embryonic stem cells in farm animals. Zygote. 1994;2(4):385-9. http://doi.org/10.1017/S0967199400002288 PMid:8665176.
    » http://doi.org/10.1017/S0967199400002288
  • Galli C, Lazzari G. 25th Anniversary of Cloning by Somatic-Cell Nuclear Transfer: current applications of SCNT in advanced breeding and genome editing in livestock. Reproduction. 2021;162(1):F23-f32. PMid:33852430.
  • Galli C, Lazzari G. Practical aspects of IVM/IVF n cattle. Anim Reprod Sci. 1996;42(1-4):371-9. http://doi.org/10.1016/0378-4320(96)01530-8
    » http://doi.org/10.1016/0378-4320(96)01530-8
  • Galli C. Animal engineering for xenotransplantation. EJT. 2023;1(3):182-91. http://doi.org/10.57603/EJT-307
    » http://doi.org/10.57603/EJT-307
  • Gandolfi F, Moor RM. Stimulation of early embryonic development in the sheep by co-culture with oviduct epithelial cells. Reproduction. 1987;81(1):23-8. http://doi.org/10.1530/jrf.0.0810023 PMid:3668954.
    » http://doi.org/10.1530/jrf.0.0810023
  • Gurdon JB. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. Development. 1962;10(4):622-40. http://doi.org/10.1242/dev.10.4.622 PMid:13951335.
    » http://doi.org/10.1242/dev.10.4.622
  • Hammer RE, Pursel VG, Rexroad CE Jr, Wall RJ, Bolt DJ, Ebert KM, Palmiter RD, Brinster RL. Production of transgenic rabbits, sheep and pigs by microinjection. Nature. 1985;315(6021):680-3. http://doi.org/10.1038/315680a0 PMid:3892305.
    » http://doi.org/10.1038/315680a0
  • Hayashi M, Zywitza V, Naitou Y, Hamazaki N, Goeritz F, Hermes R, Holtze S, Lazzari G, Galli C, Stejskal J, Diecke S, Hildebrandt TB, Hayashi K. Robust induction of primordial germ cells of white rhinoceros on the brink of extinction. Sci Adv. 2022;8(49):eabp9683. http://doi.org/10.1126/sciadv.abp9683 PMid:36490332.
    » http://doi.org/10.1126/sciadv.abp9683
  • Hayes BJ, Bowman PJ, Chamberlain AJ, Goddard ME. Invited review: Genomic selection in dairy cattle: progress and challenges. J Dairy Sci. 2009;92(2):433-43. http://doi.org/10.3168/jds.2008-1646 PMid:19164653.
    » http://doi.org/10.3168/jds.2008-1646
  • Heyman Y, Chavatte-Palmer P, Berthelot V, Fromentin G, Hocquette JF, Martignat L, Renard JP. Assessing the quality of products from cloned cattle: an integrative approach. Theriogenology. 2007;67(1):134-41. http://doi.org/10.1016/j.theriogenology.2006.09.020 PMid:17092550.
    » http://doi.org/10.1016/j.theriogenology.2006.09.020
  • Hildebrandt TB, Hermes R, Colleoni S, Diecke S, Holtze S, Renfree MB, Stejskal J, Hayashi K, Drukker M, Loi P, Goritz F, Lazzari G, Galli C. Embryos and embryonic stem cells from the white rhinoceros. Nat Commun. 2018;9(1):2589. http://doi.org/10.1038/s41467-018-04959-2 PMid:29973581.
    » http://doi.org/10.1038/s41467-018-04959-2
  • Hildebrandt TB, Holtze S, Colleoni S, Hermes R, Stejskal J, Lekolool I, Ndeereh D, Omondi P, Kariuki L, Mijele D, Mutisya S, Ngulu S, Diecke S, Hayashi K, Lazzari G, de Mori B, Biasetti P, Quaggio A, Galli C, Goeritz F. In vitro fertilization program in white rhinoceros. Reproduction. 2023;166(6):383-99. http://doi.org/10.1530/REP-23-0087 PMid:37877686.
    » http://doi.org/10.1530/REP-23-0087
  • Illmensee K, Hoppe PC. Nuclear transplantation in Mus musculus: developmental potential of nuclei from preimplantation embryos. Cell. 1981;23(1):9-18. http://doi.org/10.1016/0092-8674(81)90265-8 PMid:7214529.
    » http://doi.org/10.1016/0092-8674(81)90265-8
  • Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-21. http://doi.org/10.1126/science.1225829 PMid:22745249.
    » http://doi.org/10.1126/science.1225829
  • Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2013;14(1):49-55. http://doi.org/10.1038/nrm3486 PMid:23169466.
    » http://doi.org/10.1038/nrm3486
  • Kruip TAM, Boni R, Wurth YA, Roelofsen MWM, Pieterse MC. Potential use of ovum pick-up for embryo production and breeding in cattle. Theriogenology. 1994;42(4):675-84. http://doi.org/10.1016/0093-691X(94)90384-U PMid:16727573.
    » http://doi.org/10.1016/0093-691X(94)90384-U
  • Lander ES. The Heroes of CRISPR. Cell. 2016;164(1-2):18-28. http://doi.org/10.1016/j.cell.2015.12.041 PMid:26771483.
    » http://doi.org/10.1016/j.cell.2015.12.041
  • Lazzari G, Colleoni S, Crotti G, Turini P, Fiorini G, Barandalla M, Landriscina L, Dolci G, Benedetti M, Duchi R, Galli C. Laboratory production of equine embryos. J Equine Vet Sci. 2020;89:103097. http://doi.org/10.1016/j.jevs.2020.103097 PMid:32563445.
    » http://doi.org/10.1016/j.jevs.2020.103097
  • Lazzari G, Colleoni S, Giannelli SG, Brunetti D, Colombo E, Lagutina I, Galli C, Broccoli V. Direct derivation of neural rosettes from cloned bovine blastocysts: a model of early neurulation events and neural crest specification in vitro. Stem Cells. 2006;24(11):2514-21. http://doi.org/10.1634/stemcells.2006-0149 PMid:16931774.
    » http://doi.org/10.1634/stemcells.2006-0149
  • Lazzari G, Colleoni S, Lagutina I, Crotti G, Turini P, Tessaro I, Brunetti D, Duchi R, Galli C. Short-term and long-term effects of embryo culture in the surrogate sheep oviduct versus in vitro culture for different domestic species. Theriogenology. 2010;73(6):748-57. http://doi.org/10.1016/j.theriogenology.2009.08.001 PMid:19726075.
    » http://doi.org/10.1016/j.theriogenology.2009.08.001
  • Lazzari G, Crotti G, Turini P, Duchi R, Mari G, Zavaglia G, Barbacini S, Galli C. Equine embryos at the compacted morula and blastocyst stage can be obtained by intracytoplasmic sperm injection (ICSI) of in vitro matured oocytes with frozen-thawed spermatozoa from semen of different fertilities. Theriogenology. 2002a;58(2-4):709-12. http://doi.org/10.1016/S0093-691X(02)00777-X
    » http://doi.org/10.1016/S0093-691X(02)00777-X
  • Lazzari G, Wrenzycki C, Herrmann D, Duchi R, Kruip T, Niemann H, Galli C. Cellular and molecular deviations in bovine in vitro-produced embryos are related to the large offspring syndrome. Biol Reprod. 2002b;67(3):767-75. http://doi.org/10.1095/biolreprod.102.004481 PMid:12193383.
    » http://doi.org/10.1095/biolreprod.102.004481
  • Loi P, Ptak G, Barboni B, Fulka J Jr, Cappai P, Clinton M. Genetic rescue of an endangered mammal by cross-species nuclear transfer using post-mortem somatic cells. Nat Biotechnol. 2001;19(10):962-4. http://doi.org/10.1038/nbt1001-962 PMid:11581663.
    » http://doi.org/10.1038/nbt1001-962
  • Lonergan P, Sánchez JM. 2022. Gamete and embryo technology: multiple ovulation and embryo transfer. In: McSweeney, P.L.H., McNamara, J.P., editors. Encyclopedia of dairy sciences. 3rd ed. Oxford: Academic Press, p. 881-889. http://doi.org/10.1016/B978-0-12-818766-1.00040-4
    » http://doi.org/10.1016/B978-0-12-818766-1.00040-4
  • Lonergan P. Review: historical and futuristic developments in bovine semen technology. Animal. 2018;12(s1):s4-s18. http://doi.org/10.1017/S175173111800071X PMid:29642968.
    » http://doi.org/10.1017/S175173111800071X
  • Looney CR, Lindsey BR, Gonseth CL, Johnson DL. Commercial aspects of oocyte retrieval and in vitro fertilization (IVF) for embryo production in problem cows. Theriogenology. 1994;41(1):67-72. http://doi.org/10.1016/S0093-691X(05)80050-0
    » http://doi.org/10.1016/S0093-691X(05)80050-0
  • Matoba S, Zhang Y. Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell. 2018;23(4):471-85. http://doi.org/10.1016/j.stem.2018.06.018 PMid:30033121.
    » http://doi.org/10.1016/j.stem.2018.06.018
  • Mojica FJ, Juez G, Rodriguez-Valera F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol Microbiol. 1993;9(3):613-21. http://doi.org/10.1111/j.1365-2958.1993.tb01721.x PMid:8412707.
    » http://doi.org/10.1111/j.1365-2958.1993.tb01721.x
  • Monniaux D, Chupin D, Saumande J. Superovulatory responses of cattle. Theriogenology. 1983;19(1):55-81. http://doi.org/10.1016/0093-691X(83)90124-3
    » http://doi.org/10.1016/0093-691X(83)90124-3
  • Monniaux D, Clement F, Dalbies-Tran R, Estienne A, Fabre S, Mansanet C, Monget P. The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link? Biol Reprod. 2014;90(4):85. http://doi.org/10.1095/biolreprod.113.117077 PMid:24599291.
    » http://doi.org/10.1095/biolreprod.113.117077
  • Monniaux D. Superovulatory responses and embryo production in ruminants: lessons from ovary. In: 28th Annual Meeting A.E.T.E.; 2012 Sept 7-8; Saint Malo, France. Saint Malo: A.E.T.E.; 2012. p. 7-40.
  • Mori B, Spiriti MM, Pollastri I, Normando S, Biasetti P, Florio D, Andreucci F, Colleoni S, Galli C, Goritz F, Hermes R, Holtze S, Lazzari G, Seet S, Zwilling J, Stejskal J, Mutisya S, Ndeereh D, Ngulu S, Vigne R, Hildebrandt TB. An Ethical Assessment Tool (ETHAS) to evaluate the application of assisted reproductive technologies in mammals’ conservation: the case of the northern white rhinoceros (Ceratotherium simum cottoni). Animals (Basel). 2021;11(2):311. http://doi.org/10.3390/ani11020312 PMid:33530613.
    » http://doi.org/10.3390/ani11020312
  • Morrell JM, Keeler KD, Noakes DE, Mackenzie NM, Dresser DW. Sexing of sperm by flow cytometry. Vet Rec. 1988;122(14):322-4. http://doi.org/10.1136/vr.122.14.322 PMid:3164152.
    » http://doi.org/10.1136/vr.122.14.322
  • Niemann H, Halter R, Espanion G, Wrenzycki C, Herrmann D, Lemme E, Carnwath JW 1, Paul D. Expression of human blood clotting factor VIII (FVIII) constructs in the mammary gland of transgenic mice and sheep. J Anim Breed Genet. 1996;113(1-6):437-44. http://doi.org/10.1111/j.1439-0388.1996.tb00634.x
    » http://doi.org/10.1111/j.1439-0388.1996.tb00634.x
  • Notarianni E, Galli C, Laurie S, Moor RM, Evans MJ. Derivation of pluripotent, embryonic cell lines from the pig and sheep. J Reprod Fertil Suppl. 1991;43:255-60. PMid:1843344.
  • Palermo G, Joris H, Devroey P, Van Steirteghem AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet. 1992;340(8810):17-8. http://doi.org/10.1016/0140-6736(92)92425-F PMid:1351601.
    » http://doi.org/10.1016/0140-6736(92)92425-F
  • Pieterse MC, Kappen KA, Kruip TAM, Taverne MAM. Aspiration of bovine oocytes during transvaginal ultrasound scanning of the ovaries. Theriogenology. 1988;30(4):751-62. http://doi.org/10.1016/0093-691X(88)90310-X PMid:16726517.
    » http://doi.org/10.1016/0093-691X(88)90310-X
  • Pieterse MC, Vos PLAM, Kruip TAM, Wurth YA, van Beneden TH, Willemse AH, Taverne MAM. Transvaginal ultrasound guided follicular aspiration of bovine oocytes. Theriogenology. 1991;35(4):857-62. http://doi.org/10.1016/0093-691X(91)90426-E PMid:16726954.
    » http://doi.org/10.1016/0093-691X(91)90426-E
  • Ponsart C, Le Bourhis D, Knijn H, Fritz S, Guyader-Joly C, Otter T, Lacaze S, Charreaux F, Schibler L, Dupassieux D, Mullaart E. Reproductive technologies and genomic selection in dairy cattle. Reprod Fertil Dev. 2014;26(1):12-21. http://doi.org/10.1071/RD13328 PMid:24305173.
    » http://doi.org/10.1071/RD13328
  • Porta-Sanchez A, Mazzanti A, Tarifa C, Kukavica D, Trancuccio A, Mohsin M, Zanfrini E, Perota A, Duchi R, Hernandez-Lopez K, Jauregui-Abularach ME, Pergola V, Fernandez E, Bongianino R, Tavazzani E, Gambelli P, Memmi M, Scacchi S, Pavarino LF, Franzone PC, Lentini G, Filgueiras-Rama D, Galli C, Santiago DJ, Priori SG. Unexpected impairment of I(Na) underpins reentrant arrhythmias in a knock-in swine model of Timothy syndrome. Nat Cardiovasc Res. 2023;2(12):1291-309. http://doi.org/10.1038/s44161-023-00393-w PMid:38665938.
    » http://doi.org/10.1038/s44161-023-00393-w
  • Rizos D, Ward F, Duffy P, Boland MP, Lonergan P. Consequences of bovine oocyte maturation, fertilization or early embryo development in vitro versus in vivo: implications for blastocyst yield and blastocyst quality. Mol Reprod Dev. 2002;61(2):234-48. http://doi.org/10.1002/mrd.1153 PMid:11803560.
    » http://doi.org/10.1002/mrd.1153
  • Robertson E, Bradley A, Kuehn M, Evans M. Germ-line transmission of genes introduced into cultured pluripotential cells by retroviral vector. Nature. 1986;323(6087):445-8. http://doi.org/10.1038/323445a0 PMid:3762693.
    » http://doi.org/10.1038/323445a0
  • Staigmiller R, Moor R. Effect of follicle cells on the maturation and developmental competence of ovine oocytes matured outside the follicle. Gamete Res. 1984;9(2):221-9. http://doi.org/10.1002/mrd.1120090211
    » http://doi.org/10.1002/mrd.1120090211
  • Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978;312(8085):366. http://doi.org/10.1016/S0140-6736(78)92957-4 PMid:79723.
    » http://doi.org/10.1016/S0140-6736(78)92957-4
  • Sung LY, Gao S, Shen H, Yu H, Song Y, Smith SL, Chang CC, Inoue K, Kuo L, Lian J, Li A, Tian XC, Tuck DP, Weissman SM, Yang X, Cheng T. Differentiated cells are more efficient than adult stem cells for cloning by somatic cell nuclear transfer. Nat Genet. 2006;38(11):1323-8. http://doi.org/10.1038/ng1895 PMid:17013394.
    » http://doi.org/10.1038/ng1895
  • Thibier M, Nibart M. The sexing of bovine embryos in the field. Theriogenology. 1995;43(1):71-80. http://doi.org/10.1016/0093-691X(94)00008-I
    » http://doi.org/10.1016/0093-691X(94)00008-I
  • Thibier M. The European embryo transfer industry in cattle - a challange in 1984 - a success in 2014 -and well supported and reported by the AETE. In: Proceedings of the 30th Scientific meeting of A.E.T.E.; 2014 Sept 12-13; Dresden, Germany. Dresden: A.E.T.E.; 2014. p. 1-13.
  • Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145-7. http://doi.org/10.1126/science.282.5391.1145 PMid:9804556.
    » http://doi.org/10.1126/science.282.5391.1145
  • Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11(9):636-46. http://doi.org/10.1038/nrg2842 PMid:20717154.
    » http://doi.org/10.1038/nrg2842
  • Vos PL, van der Schans A, de Wit AA, Bevers MM, Willemse AH, Dieleman SJ. Effects of neutralization of pregnant mares’ serum gonadotrophin (PMSG) shortly before or at the preovulatory LH surge in PMSG-superovulated heifers on follicular function and development. Reproduction. 1994;100(2):387-93. http://doi.org/10.1530/jrf.0.1000387 PMid:8021854.
    » http://doi.org/10.1530/jrf.0.1000387
  • Willadsen SM. Nuclear transplantation in sheep embryos. Nature. 1986;320(6057):63-5. http://doi.org/10.1038/320063a0 PMid:3951549.
    » http://doi.org/10.1038/320063a0
  • Wilmut I, Rowson LEA. Experiments on the low-temperature preservation of cow embryos. Vet Rec. 1973;92(26):686-90. http://doi.org/10.1136/vr.92.26.686 PMid:4730118.
    » http://doi.org/10.1136/vr.92.26.686
  • Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997;385(6619):810-3. http://doi.org/10.1038/385810a0 PMid:9039911.
    » http://doi.org/10.1038/385810a0
  • Wilson JM, Jones AL, Moore K, Looney CR, Bondioli KR. Superovulation of cattle with a recombinant-DNA bovine follicle stimulating hormone. Anim Reprod Sci. 1993;33(1-4):71-82. http://doi.org/10.1016/0378-4320(93)90107-3
    » http://doi.org/10.1016/0378-4320(93)90107-3
  • Wright JM. Non-surgical embryo transfer in cattle embryo-recipient interactions. Theriogenology. 1981;15(1):43-56. http://doi.org/10.1016/S0093-691X(81)80017-9 PMid:16725539.
    » http://doi.org/10.1016/S0093-691X(81)80017-9
  • Yang X, Smith SL, Tian XC, Lewin HA, Renard JP, Wakayama T. Nuclear reprogramming of cloned embryos and its implications for therapeutic cloning. Nat Genet. 2007;39(3):295-302. http://doi.org/10.1038/ng1973 PMid:17325680.
    » http://doi.org/10.1038/ng1973
  • Young LE, Sinclair KD, Wilmut I. Large offspring syndrome in cattle and sheep. Rev Reprod. 1998;3(3):155-63. http://doi.org/10.1530/ror.0.0030155 PMid:9829550.
    » http://doi.org/10.1530/ror.0.0030155
  • Zhou Q, Renard JP, Le Friec G, Brochard V, Beaujean N, Cherifi Y, Fraichard A, Cozzi J. Generation of fertile cloned rats by regulating oocyte activation. Science. 2003;302(5648):1179. http://doi.org/10.1126/science.1088313 PMid:14512506.
    » http://doi.org/10.1126/science.1088313
  • Zywitza V, Rusha E, Shaposhnikov D, Ruiz-Orera J, Telugu N, Rishko V, Hayashi M, Michel G, Wittler L, Stejskal J, Holtze S, Göritz F, Hermes R, Wang J, Izsvák Z, Colleoni S, Lazzari G, Galli C, Hildebrandt TB, Hayashi K, Diecke S, Drukker M. Naïve-like pluripotency to pave the way for saving the northern white rhinoceros from extinction. Sci Rep. 2022;12(1):3100. http://doi.org/10.1038/s41598-022-07059-w PMid:35260583.
    » http://doi.org/10.1038/s41598-022-07059-w

Publication Dates

  • Publication in this collection
    26 Aug 2024
  • Date of issue
    2024

History

  • Received
    07 May 2024
  • Accepted
    20 June 2024
Colégio Brasileiro de Reprodução Animal Coronel José dias Bicalho, 1224, CEP: , 31275-050, Belo Horizonte, MG - Brasil, Tel.: 55-31-3491 7122 - Belo Horizonte - MG - Brazil
E-mail: animreprod.journal@gmail.com