Acessibilidade / Reportar erro

40 ‘wild’ years: the current reality and future potential of assisted reproductive technologies in wildlife species

Abstract

Over the past 40 years, assisted reproductive technologies (ARTs) have grown significantly in scale and innovation, from the bovine embryo industry’s shift from in vivo derived to in vitro produced embryos and the development of somatic cell-based approaches for embryo production. Domestic animal models have been instrumental in the development of ARTs for wildlife species in support of the One Plan Approach to species conservation that integrates in situ and ex situ population management strategies. While ARTs are not the sole solution to the biodiversity crisis, they can offer opportunities to maintain, and even improve, the genetic composition of the captive and wild gene pools over time. This review focuses on the application of sperm and embryo technologies (artificial insemination and multiple ovulation/in vitro produced embryo transfer, respectively) in wildlife species, highlighting impactful cases in which significant progress or innovation has transpired. One of the key messages following decades of efforts in this field is the importance of collaboration between researchers and practitioners from zoological, academic, governmental, and private sectors.

Keywords:
assisted reproduction; embryo technologies; biodiversity conservation; wildlife species

Introduction

Decreasing wildlife population trends are coupled with concerns of ecosystem disruption that have far-reaching impact on animals and humans alike. The International Union for Conservation of Nature Species Survival Commission’s (IUCN SSC’s) One Plan Approach to conservation promotes in situ and ex situ connectivity in population management to enhance species sustainability goals (Byers et al., 2013Byers O, Lees C, Wilcken J, Schwitzer C. The One Plan Approach: the philosophy and implementation of CBSG’s approach to integrated species conservation planning. In: Gusset M, Dick G, editors. Towards integrated species conservation. Vol. 14. Switzerland: World Association of Zoos and Aquariums Magazine. [serial on the internet]. 2013 [cited 2024 March 9]. p. 2-5. Available from: https://www.waza.org/wp-content/uploads/2019/02/waza_mag_14.pdf.
https://www.waza.org/wp-content/uploads/...
). Assisted reproductive technologies (ARTs) can have a significant impact on achieving species recovery targets through long-term storage of genetic material as assurance against on-going or future depletion of diversity. While ARTs are not the miracle solution for fighting extinction, they are a powerful supporting tool for genetic management across time. Interest in the application of ARTs in wildlife species began in the mid-1970s with the transfers of flushed ‘exotic’ embryos into recipients of related livestock species (Kydd et al., 1985Kydd J, Boyle MS, Allen WR, Shephard A, Summer PM. Transfer of exotic equine embryos to domestic horses and donkeys. Equine Vet J. 1985;17(S3):80-3. http://doi.org/10.1111/j.2042-3306.1985.tb04601.x.
http://doi.org/10.1111/j.2042-3306.1985....
; Dresser, 1986Dresser BL. Embryo transfer in exotic bovids. Int Zoo Yearb. 1986;24(1):138-42. http://doi.org/10.1111/j.1748-1090.1985.tb02529.x.
http://doi.org/10.1111/j.1748-1090.1985....
). They gained momentum in the mid-1980s with the landmark paper by Jonathan Ballou, a researcher focused on the genetic and demographic challenges of small population management (Ballou, 1984Ballou JD. Strategies for maintaining genetic diversity in captive populations through reproductive technology. Zoo Biol. 1984;3(4):311-23. http://doi.org/10.1002/zoo.1430030404.
http://doi.org/10.1002/zoo.1430030404...
). The wide range of reproductive strategies across vertebrate taxa (see Figure 1 for sperm and oocyte diversity) significantly influenced the development of ART programs, and the importance of domestic animal models was recognized early on (Wildt et al., 1986Wildt DE, Schiewe MC, Schmidt PM, Goodrowe KL, Howard JG, Phillips LG, O’Brien SJ, Bush M. Developing animal model systems for embryo technologies in rare and endangered wildlife. Theriogenology. 1986;25(1):33-51. http://doi.org/10.1016/0093-691X(86)90182-2.
http://doi.org/10.1016/0093-691X(86)9018...
). This review will touch briefly on the progress of artificial insemination (AI) as a conservation breeding tool and focus on the potential for embryo technologies, specifically multiple ovulation embryo transfer (MOET) and in vitro produced embryo transfer (IVPET), to meet the growing threats to species sustainability.

Figure 1
Diversity in gamete morphology across vertebrate taxa. Stylized sperm and egg drawings depict broad similarities and differences among Chondrichthyes, Osteichthyes, Amphibia, Reptilia, Aves, and Mammalia. There are further species-specific differences that are not captured in this schematic. (A) proportion of sperm head, midpiece and tail lengths among vertebrate classes. Total sperm lengths range from 20 – 350 µm depending on the species. Ext. = external; only sperm morphology from external (and not internal) fertilizers are shown. Sperm figure modified from Kahrl et al. (2022)Kahrl AF, Snook RR, Fitzpatrick JL. Fertilization mode differentially impacts the evolution of vertebrate sperm components. Nat Commun. 2022;13(1):6809. http://doi.org/10.1038/s41467-022-34609-7. PMid:36357384.
http://doi.org/10.1038/s41467-022-34609-...
by removing sperm images from internal fertilizers for Osteichthyes and Amphibia; licensed under a Creative Commons Attribution 4.0 International License (CC BY 4.0; https://creativecommons.org/.licenses/by/4.0/); (B) cellular and extracellular layers encompassing the eggs (or oocytes) of different vertebrate classes (Menkhorst and Selwood, 2008Menkhorst E, Selwood L. Vertebrate extracellular preovulatory and postovulatory egg coats. Biol Reprod. 2008;79(5):790-7. http://doi.org/10.1095/biolreprod.108.068551. PMid:18614703.
http://doi.org/10.1095/biolreprod.108.06...
). Egg diameters range from 100 – 100,000 µm depending on the species. Egg images were created with BioRender.com (agreement number TG26OWIEYQ). Silhouette illustrations were contributed by various authors under public domain license (CC0 1.0 license) from PhyloPic (http://phylopic.org).

Sperm Technologies: Impact of Artificial Insemination

The past century demonstrated the potential of sperm-focused technologies. In wildlife species, insemination of females under natural or stimulated cycles continues to be the most effective ART for offspring production. To date, artificial insemination (AI) has been attempted successfully in ~100 species across diverse taxa (Swanson and Penfold, 2018Swanson WF, Penfold LM. Captive wildlife breeding: mammals & birds. In: Skinner MK, editor. Encyclopedia of Reproduction. 2nd ed. Cambridge: Academic Press; 2018. p. 782-90. http://doi.org/10.1016/B978-0-12-809633-8.20631-6.
http://doi.org/10.1016/B978-0-12-809633-...
) with reptiles just beginning to benefit from an increased investment in the development of ARTs (Perry, 2021Perry S. Developing assisted reproduction for reptiles, what’s next? Clin Theriogenology. 2021;13:383. https://doi.org/10.58292/ct.v13.9315.). Protocols for external fertilizers, such as fishes and amphibians, have also advanced, and will be discussed in the section below on in vitro embryo production. The power of AI stems from its widespread application due to the lower investment of resources (funds, specialized equipment, technical expertise), but also because the embryo is developing in its natural environment, precluding the need for in-depth knowledge of pre-implantation embryo development.

Sperm from wild mammals is collected by electroejaculation or post-mortem epididymal dissection, and more recently by urethral catheterization (Lueders et al., 2012Lueders I, Luther I, Scheepers G, van der Horst G. Improved semen collection method for wild felids: urethral catheterization yields high sperm quality in African lions (Panthera leo). Theriogenology. 2012;78(3):696-701. http://doi.org/10.1016/j.theriogenology.2012.02.026. PMid:22538007.
http://doi.org/10.1016/j.theriogenology....
; Prieto et al., 2014Prieto MT, Sanchez-Calabuig MJ, Hildebrandt TB, Santiago-Moreno J, Saragusty J. Sperm cryopreservation in wild animals. Eur J Wildl Res. 2014;60(6):851-64. http://doi.org/10.1007/s10344-014-0858-4.
http://doi.org/10.1007/s10344-014-0858-4...
). The latter approach will be beneficial as animal welfare guidelines continue to change, making minimally invasive alternatives a necessity. In non-mammalian species, sperm samples are obtained by massage, artificial vagina, or catheterization (reviewed by Prieto et al., 2014Prieto MT, Sanchez-Calabuig MJ, Hildebrandt TB, Santiago-Moreno J, Saragusty J. Sperm cryopreservation in wild animals. Eur J Wildl Res. 2014;60(6):851-64. http://doi.org/10.1007/s10344-014-0858-4.
http://doi.org/10.1007/s10344-014-0858-4...
). For many mammals, birds and fishes, sperm handling solutions are commercially available from related domestic animals and humans. In species where a commercial sperm extender is unavailable or not providing the desired outcomes, modification of a commercial product (e.g., by altering glycerol concentration) or optimization of a ‘home-made’ recipe is required.

Fresh/chilled sperm has produced viable offspring across all taxa (reviewed by Blanco et al. (2009)Blanco JM, Wildt DE, Höfle U, Voelker W, Donoghue AM. Implementing artificial insemination as an effective tool for ex situ conservation of endangered avian species. Theriogenology. 2009;71(1):200-13. http://doi.org/10.1016/j.theriogenology.2008.09.019. PMid:19004491.
http://doi.org/10.1016/j.theriogenology....
(birds); Kouba et al. (2009)Kouba AJ, Vance CK, Willis EL. Artificial fertilization for amphibian conservation: current knowledge and future considerations. Theriogenology. 2009;71(1):214-27. http://doi.org/10.1016/j.theriogenology.2008.09.055. PMid:19026442.
http://doi.org/10.1016/j.theriogenology....
(amphibians); Beirão et al. (2019)Beirão J, Boulais M, Gallego V, O’Brien JK, Peixoto S, Robeck TR, Cabrita E. Sperm handling in aquatic animals for artificial reproduction. Theriogenology. 2019;133:161-78. http://doi.org/10.1016/j.theriogenology.2019.05.004. PMid:31108371.
http://doi.org/10.1016/j.theriogenology....
(fishes); Mastromonaco and Songsasen (2020)Mastromonaco GF, Songsasen N. Reproductive technologies for the conservation of wildlife and endangered species. In: Presicce GA, editor. Reproductive technologies in animals. Cambridge: Academic Press; 2020. Chapter 7, p. 99-117. (mammals/birds); Perry (2021)Perry S. Developing assisted reproduction for reptiles, what’s next? Clin Theriogenology. 2021;13:383. https://doi.org/10.58292/ct.v13.9315. (reptiles)); however, implementation of ARTs for long-term genetic management requires access to sperm over extended periods of time for the infusion of novel or minimally represented genes. As a result, sperm cryobiology has been an active field of study resulting in hundreds of published papers detailing the effects on pre- and post-thaw sperm characteristics. Sperm cryopreservation and use in ARTs have been successful in many of the species attempted, even if just a one-time success. Species exhibiting chilling sensitivity traits can be challenging despite extensive efforts to optimize species-specific techniques. In these cases, enhancement of standard protocols may provide additional protection to sperm membranes to improve post-thaw motility and integrity, such as the use of specialized density gradients or oviductal extracellular vesicles in cheetahs (Acinonyx jubatus) (Crosier et al., 2009Crosier AE, Henghali JN, Howard J, Pukazhenthi BS, Terrell KA, Marker LL, Wildt DE. Improved quality of cryopreserved cheetah (Acinonyx jubatus) spermatozoa after centrifugation through Accudenz. J Androl. 2009;30(3):298-308. http://doi.org/10.2164/jandrol.108.006239. PMid:19023140.
http://doi.org/10.2164/jandrol.108.00623...
; Ferraz et al., 2020Ferraz MAMM, Nagashima JB, Noonan MJ, Crosier AE, Songsasen N. Oviductal extracellular vesicles improve post-thaw sperm function in red wolves and cheetahs. Int J Mol Sci. 2020;21(10):3733. http://doi.org/10.3390/ijms21103733. PMid:32466321.
http://doi.org/10.3390/ijms21103733...
). More recently, several researchers shifted their focus to freeze-drying sperm, and preliminary trials showed promising outcomes (Kaneko et al., 2014Kaneko T, Ito H, Sakamoto H, Onuma M, Inoue-Murayama M. Sperm preservation by freeze-drying for the conservation of wild animals. PLoS One. 2014;9(11):e113381. http://doi.org/10.1371/journal.pone.0113381. PMid:25409172.
http://doi.org/10.1371/journal.pone.0113...
; Anzalone et al., 2018Anzalone DA, Palazzese L, Iuso D, Martino G, Loi P. Freeze-dried spermatozoa: an alternative biobanking option for endangered species. Anim Reprod Sci. 2018;190:85-93. http://doi.org/10.1016/j.anireprosci.2018.01.010. PMid:29397252.
http://doi.org/10.1016/j.anireprosci.201...
). The possibility of eliminating the dependence on liquid nitrogen makes this an attractive method for field collections and resource-restricted conservation biobanks.

Insemination of naturally cycling or hormonally primed females using either fresh, chilled or frozen-thawed sperm has been accomplished through a variety of methods: non-surgically using transcervical (mammals) / transcloacal (birds, reptiles) sperm deposition, or surgically using laparotomic or laparoscopic sperm deposition into the uterine cavity or oviduct (Swanson and Penfold, 2018Swanson WF, Penfold LM. Captive wildlife breeding: mammals & birds. In: Skinner MK, editor. Encyclopedia of Reproduction. 2nd ed. Cambridge: Academic Press; 2018. p. 782-90. http://doi.org/10.1016/B978-0-12-809633-8.20631-6.
http://doi.org/10.1016/B978-0-12-809633-...
). Notably, for AI to be successful, the female must be at the right stage of the ovarian cycle to receive the sperm, except perhaps in species with long-term sperm storage capabilities within the female (e.g., bats: 6 months; lizards: up to 1.5 years; sharks: 1-2 years; snakes: up to 7 years; Holt and Fazeli (2016)Holt WV, Fazeli A. Sperm storage in the female reproductive tract. Annu Rev Anim Biosci. 2016;4(1):291-310. http://doi.org/10.1146/annurev-animal-021815-111350 PMid:26526545.
http://doi.org/10.1146/annurev-animal-02...
. The important topic of ovarian stimulation will be discussed further in the embryo technologies section below.

One of the greatest challenges for wildlife ARTs is advancing the technique from research to application. With limited access to research animals and materials, protocol development and implementation can take multiple decades. Thus, many of the AI births to date have been proof-of-concept attempts that generated media attention without further progress. The slow advancement and relatively low success rates present a significant challenge when advocating for the inclusion of ARTs in conservation breeding programs. Despite these drawbacks, AI has been used not only to achieve ex situ targets (sustainability of captive populations), but also to support in situ needs (recovery of the species in their native ranges).

Spotlight: Common Bottlenose Dolphin (Tursiops truncatus)

Throughout the 2000s, Todd Robeck and Justine O’Brien spear-headed a comprehensive sperm-based approach that included fresh-chilled, frozen-thawed and sex-sorted sperm inseminations in bottlenose dolphins. AI offered an opportunity to overcome the challenges of managing the genetics and demographics of a closed captive population in a species that is difficult to translocate between institutions. Ovarian synchronization, manual semen collection and sperm cryopreservation protocols were developed, and intra-uterine inseminations resulted in 60-70% pregnancy rates for fresh-chilled, frozen-thawed un-sorted and frozen-thawed sex-sorted sperm (O’Brien and Robeck, 2006O’Brien JK, Robeck TR. Development of sperm sexing and associated assisted reproductive technology for sex preselection of captive bottlenose dolphins (Tursiops truncatus). Reprod Fertil Dev. 2006;18(3):319-29. http://doi.org/10.1071/RD05108. PMid:16554007.
http://doi.org/10.1071/RD05108...
; Robeck et al., 2013Robeck TR, Montano GA, Steinman KJ, Smolensky P, Sweeney J, Osborn S, O’Brien JK. Development and evaluation of deep intra-uterine artificial insemination using cryopreserved sexed spermatozoa in bottlenose dolphins (Tursiops truncatus). Anim Reprod Sci. 2013;139(1-4):168-81. http://doi.org/10.1016/j.anireprosci.2013.04.004. PMid:23660366.
http://doi.org/10.1016/j.anireprosci.201...
). To date, 30 bottlenose dolphin calves have been born from AI using sex-sorted frozen-thawed sperm with a sex predetermination rate of 93% (United Parks & Resorts, 2024United Parks & Resorts. [homepage on the Internet]. United Parks & Resorts; 2024. Species preservation laboratory; 2024 [cited 2024 March 9]. Available from: https://seaworld.org/conservation/species-preservation-laboratory/primary-research/.
https://seaworld.org/conservation/specie...
), making it one of the greatest achievements in wildlife AI. Building on this work, successful inseminations have resulted in pacific white-sided dolphin (Lagenorhynchus obliquidens; Robeck et al., 2009Robeck TR, Steinman KJ, Greenwell M, Ramirez K, Van Bonn W, Yoshioka M, Katsumata E, Dalton L, Osborn S, O’Brien JK. Seasonality, estrous cycle characterization, estrus synchronization, semen cryopreservation, and artificial insemination in the Pacific white-sided dolphin (Lagenorhynchus obliquidens). Reproduction. 2009;138(2):391-405. http://doi.org/10.1530/REP-08-0528. PMid:19494046.
http://doi.org/10.1530/REP-08-0528...
), orca (Orcinus orca; Robeck et al., 2004Robeck TR, Steinman KJ, Gearhart S, Reidarson TR, McBain JF, Monfort SL. Reproductive physiology and development of artificial insemination technology in killer whales (Orcinus orca). Biol Reprod. 2004;71(2):650-60. http://doi.org/10.1095/biolreprod.104.027961. PMid:15115725.
http://doi.org/10.1095/biolreprod.104.02...
), and beluga whale calves (Delphinapterus leucas; O’Brien et al., 2008O’Brien JK, Steinman KJ, Schmitt T, Robeck TR. Semen collection, characterisation and artificial insemination in the beluga (Delphinapterus leucas) using liquid-stored spermatozoa. Reprod Fertil Dev. 2008;20(7):770-83. http://doi.org/10.1071/RD08031. PMid:18842179.
http://doi.org/10.1071/RD08031...
). Although ART development has primarily benefitted marine mammals in managed care, the groundwork has been laid for possible application in future wild cetacean recovery efforts.

Embryo Technologies: Enhanced Potential for Genetic Rescue

With the on-going pressure to preserve and restore biodiversity, there is an urgency to include female genetics in the wildlife biobanks. This requires investment in embryo technologies, specifically the acquisition of a) oocytes for fertilization or cryopreservation, b) embryos for transfer or cryopreservation, and c) somatic cells for production of gametes and embryos. These techniques are dependent on species-specific knowledge of the female’s reproductive biology and the embryo’s early development. Despite the additional challenges, the need to ensure female genetic contribution to conservation populations has stimulated concentrated efforts in various threatened species.

The 1970s and 80s sparked an interest in interspecific transfers to investigate the possibility of using domestic animal surrogates to gestate endangered species embryos. These early attempts included the transfers of in vivo derived (IVD) embryos from Grant’s zebra (Equus quagga boehmi) and Przewalski’s horse (Equua ferus przewalskii) into domestic mare, bongo antelope (Tragelaphus eurycerus) into common eland antelope (Taurotragus oryx), and more (Dresser, 1988Dresser BL. Cryobiology, embryo transfer, and artificial insemination in ex situ animal conservation programs. In: Wilson EO, Peter FM, editors. Biodiversity. Washington (DC): National Academies Press; 1988. Chapter 34, p. 296-308.; Allen and Wilsher, 2020Allen WRT, Wilsher S. Historical aspects of equine embryo transfer. J Equine Vet Sci. 2020;89:102987. http://doi.org/10.1016/j.jevs.2020.102987. PMid:32563443.
http://doi.org/10.1016/j.jevs.2020.10298...
). The subsequent decades brought a re-focusing of attention onto basic reproductive biology to expand the knowledge of reproductive anatomy, physiology, and gamete biology (Pukazhenthi and Wildt, 2004Pukazhenthi BS, Wildt DE. Which reproductive technologies are most relevant to studying, managing and conserving wildlife? Reprod Fertil Dev. 2004;16(1-2):33-46. http://doi.org/10.1071/RD03076. PMid:14972101.
http://doi.org/10.1071/RD03076...
; Herrick, 2019Herrick JR. Assisted reproductive technologies for endangered species conservation: developing sophisticated protocols with limited access to animals with unique reproductive mechanisms. Biol Reprod. 2019;100(5):1158-70. http://doi.org/10.1093/biolre/ioz025. PMid:30770538.
http://doi.org/10.1093/biolre/ioz025...
). Thus, while live births from embryo technologies have been documented, the outcomes have not been as consistent and widespread as AI, apart from fishes and amphibians. Currently, embryo technologies are not being implemented as a population management strategy in mammals, birds, or reptiles.

  1. Ovarian Cycle Monitoring and Control

The female component of ARTs requires monitoring or control of ovarian dynamics in the oocyte donors and embryo recipients. For offspring-endpoint AIs, both natural and ‘artificially’ synchronized (i.e., application of exogenous hormones or alteration of environmental conditions such as changes in temperature or humidity) cycles are used. When using natural cycles, as in the giant panda, non-invasive hormone monitoring (urine and feces) is a necessity since blood sampling and ultrasonography are not possible in species that cannot be routinely handled (Kersey et al., 2010Kersey DC, Wildt DE, Brown JL, Snyder RJ, Huang Y, Monfort SL. Endocrine milieu of perioestrus in the giant panda (Ailuropoda melanoleuca), as determined by non-invasive hormone measures. Reprod Fertil Dev. 2010;22(6):901-12. http://doi.org/10.1071/RD09178. PMid:20591324.
http://doi.org/10.1071/RD09178...
). In contrast, many elephants are trained for blood collection allowing serum LH levels to be used to time inseminations during natural cycles, a feat that is possible only due to the double LH peaks in African and Asian elephants (Loxodonta africana and Elaphas maximus, respectively; Thitaram and Brown, 2018Thitaram C, Brown JL. Monitoring and controlling ovarian activity in elephants. Theriogenology. 2018;109:42-7. http://doi.org/10.1016/j.theriogenology.2017.12.009. PMid:29287954.
http://doi.org/10.1016/j.theriogenology....
). Aside from the species that can be trained for blood sampling, the role of non-invasive hormone monitoring in conservation breeding programs cannot be underestimated. Fecal hormone profiles have helped shed some light on the complexities of embryonic diapause, follicular stasis, and more. Similarly, the use of transrectal ultrasound in animals that can be chute trained has been instrumental in elucidating ovarian dynamics and establishing the timing for inseminations and embryo transfers (Pennington et al., 2019Pennington PM, Marshall KL, Capiro JM, Felton RG, Durrant BS. Ovulation induction in anovulatory southern white rhinoceros (Ceratotherium simum simum) without altrenogest. Conserv Physiol. 2019;7(1):coz033. http://doi.org/10.1093/conphys/coz033. PMid:31249689.
http://doi.org/10.1093/conphys/coz033...
).

Ovarian synchronization with exogenous hormones has been successfully applied in many mammalian species using protocols generally adopted from farm and laboratory animals with certain species-specific modifications to optimize outcomes. In wild felids, gonadotropin dosages based on domestic cat studies proved difficult to extrapolate to diverse wild cat species on size alone due to species-specific differences in sensitivities to eCG and hCG: ocelots (Leopardus pardalis; 9 kg) required double the dose than cheetahs (35 kg), whereas clouded leopards (Neofelis nebulosa; 15 kg) required the same dose as domestic cats (2 kg) (Thongphakdee et al., 2018Thongphakdee A, Tipkantha W, Punkong C, Chatdarong K. Monitoring and controlling ovarian activity in wild felids. Theriogenology. 2018;109:14-21. http://doi.org/10.1016/j.theriogenology.2017.12.010. PMid:29258698.
http://doi.org/10.1016/j.theriogenology....
). Studies in domestic cats and cheetahs demonstrated that progestin priming prior to gonadotropin administration normalized the endocrine response thereby improving ovulation, corpus luteum function and oocyte developmental potential (Stewart et al., 2012Stewart RA, Pelican KM, Crosier AE, Pukazhenthi BS, Wildt DE, Ottinger MA, Howard J. Oral progestin priming increases ovarian sensitivity to gonadotropin stimulation and improves luteal function in the cat. Biol Reprod. 2012;87(6):137. http://doi.org/10.1095/biolreprod.112.104190. PMid:23100619.
http://doi.org/10.1095/biolreprod.112.10...
; Crosier et al., 2017Crosier AE, Comizzoli P, Koester DC, Wildt DE. Circumventing the natural, frequent oestrogen waves of the female cheetah (Acinonyx jubatus) using oral progestin (Altrenogest). Reprod Fertil Dev. 2017;29(8):1486-98. http://doi.org/10.1071/RD16007. PMid:27483379.
http://doi.org/10.1071/RD16007...
).

Acquisition of multiple oocytes from donor females requires ovarian super-stimulation in monovulatory species, as well as certain polyovulatory species. Application of exogenous hormones (e.g., FSH) to enhance follicular recruitment must maintain a balance between producing a maximum number of follicles and ensuring the retrieval of competent oocytes. Domestic cattle hormone regimens have formed the basis of many wildlife super-stimulation trials, ranging from wild bovids (e.g., banteng (Bos javanicus)) to antelopes (e.g., addax (Addax nasomaculatus)) (Sontakke, 2018Sontakke SD. Monitoring and controlling ovarian activities in wild ungulates. Theriogenology. 2018;109:31-41. http://doi.org/10.1016/j.theriogenology.2017.12.008. PMid:29254686.
http://doi.org/10.1016/j.theriogenology....
). Notably, studies in domestic cattle demonstrated that extrapolation between even the more closely related species and subspecies, namely holstein (Bos taurus), gir (Bos indicus), and murrah cattle (Bubalus bubalis), is problematic due to the inherent differences in follicular waves, antral follicle populations, and follicle and corpus luteum diameters (Baldrighi et al., 2022Baldrighi JM, Sá Filho MF, Siqueira AF, Visintin JA, Baruselli PS, Assumpção MEOA. Temporal evaluation of follicular dynamics and endocrine patterns of Holstein (Bos taurus), Gir (Bos indicus), and Murrah (Bubalus bubalis) heifers kept under the same nutritional, management and environmental conditions. Theriogenology. 2022;190:8-14. http://doi.org/10.1016/j.theriogenology.2022.07.006. PMid:35863098.
http://doi.org/10.1016/j.theriogenology....
).

For non-mammalian species, similar approaches employing exogenous hormones have been documented. In amphibians, both spermiation and ovulation can be induced with injections of hCG or GnRH; however, in certain species such as the Puerto Rican crested toad, hormones alone were not effective in stimulating ovulation, and altered environmental conditions (e.g., hibernation at low temperatures) were required prior to hormone administration to enhance outcomes (Kouba and Vance, 2009Kouba AJ, Vance CK. Applied reproductive technologies and genetic resource banking for amphibian conservation. Reprod Fertil Dev. 2009;21(6):719-37. http://doi.org/10.1071/RD09038. PMid:19567216.
http://doi.org/10.1071/RD09038...
). In both fishes and amphibians, addition of dopamine antagonists to GnRH protocols improved ovulation and fertilization (Van Eenennaam et al., 2008Van Eenennaam JP, Linares-Casenave J, Muguet J-B, Doroshov SI. Induced spawning, artificial fertilization, and egg incubation techniques for green sturgeon. N Am J Aquaculture. 2008;70(4):434-45. http://doi.org/10.1577/A07-073.1.
http://doi.org/10.1577/A07-073.1...
; Silla et al., 2021Silla AJ, Calatayud NE, Trudeau VL. Amphibian reproductive technologies: approaches and welfare considerations. Conserv Physiol. 2021;9(1):coab011. http://doi.org/10.1093/conphys/coab011. PMid: 33763231.
http://doi.org/10.1093/conphys/coab011...
). In birds, on the other hand, inseminations have been timed to natural laying cycles (Swengel and Tuite, 1997Swengel SR, Tuite ML. Recent advances in scheduling strategies and practical techniques in crane artificial insemination. In: North American Crane Workshop Proceedings; 1997 Jan 10-13; Biloxi, United States. Biloxi: North American Crane; 1997. p. 46-55.), with alterations in photoperiod being used to induce egg laying outside of the breeding season (Zhu et al., 2017Zhu H, Shao X, Chen Z, Wei C, Lei M, Ying S, Yu J, Shi Z. Induction of out-of-season egg laying by artificial photoperiod in Yangzhou geese and the associated endocrine and molecular regulation mechanisms. Anim Reprod Sci. 2017;180:127-36. http://doi.org/10.1016/j.anireprosci.2017.03.009. PMid:28342689.
http://doi.org/10.1016/j.anireprosci.201...
).

  1. Multiple Ovulation Embryo Transfer (MOET)

MOET has not been widely attempted in wild mammals. Retrieval of multiple embryos from naturally bred or artificially inseminated donor females are either transferred fresh to recipient females or cryopreserved for future use; an effective method for increasing a female’s lifetime reproductive output. Despite the abundance of data from domestic cattle studies highlighting the increased competence of IVD embryos compared to in vitro produced (IVP) embryos (Ferré et al., 2020Ferré LB, Kjelland ME, Strøbech LB, Hyttel P, Mermillod P, Ross PJ. Review: recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods. Animal. 2020;14(5):991-1004. http://doi.org/10.1017/S1751731119002775. PMid:31760966.
http://doi.org/10.1017/S1751731119002775...
), wildlife researchers interested in obtaining female genetics have focused on IVPET. Thus, aside from the early attempts on interspecies ET and efforts in species of commercial importance (e.g., dromedary camels (Camelus dromedarius), red deer (Cervus elaphus)), MOET has been applied in only a handful of species of socio-cultural, economic, and nutritional relevance to indigenous communities (wood bison (Bison bison athabascae), yak (Bos grunniens), llama (Lama glama), alpaca (Vicugna pacos); Mastromonaco, 2024Mastromonaco GF. A quarter century of CANDES: the state of embryo technologies in companion animals, non-domestic and endangered species. Theriogenology Wild. 2024;4:100069. http://doi.org/10.1016/j.therwi.2023.100069.
http://doi.org/10.1016/j.therwi.2023.100...
). The lack of traction with MOET in conservation breeding programs has been due to difficulties in accessing and handling invaluable donor females. However, even in domestic cattle, there has been a shift away from MOET towards IVPET, with the number of transferable IVP embryos now surpassing the number of IVD embryos produced annually; a change that corresponds with the increasing efficiency of IVP systems (Ferré et al., 2020Ferré LB, Kjelland ME, Strøbech LB, Hyttel P, Mermillod P, Ross PJ. Review: recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods. Animal. 2020;14(5):991-1004. http://doi.org/10.1017/S1751731119002775. PMid:31760966.
http://doi.org/10.1017/S1751731119002775...
).

Spotlight: Wood Bison (Bison bison athabascae)

Since 2006, Gregg Adams has led a systematic plan to establish a biobank of disease-free sperm and embryos from free-ranging wood bison to mitigate the effects of tuberculosis and brucellosis in wild herds (Bison Integrated Genomics (BIG) Project, 2022Bison Integrated Genomics (BIG) Project. [homepage on the Internet]. Parks Canada; 2022 [cited 2024 March 9]. Available from: https://www.bigproject.ca/.
https://www.bigproject.ca/...
). Domestic cattle synchronization and super-stimulation protocols have been somewhat effective in wood bison, particularly with modifications (i.e., single slow-release FSH dose) to reduce handling frequency and stress (Toosi et al., 2013Toosi BM, Tribulo A, Lessard C, Mastromonaco GF, McCorkell RB, Adams GP. Superovulation and embryo transfer in wood bison (Bison bison athabascae). Theriogenology. 2013;80(5):542-51. http://doi.org/10.1016/j.theriogenology.2013.05.020. PMid:23831114.
http://doi.org/10.1016/j.theriogenology....
). Interestingly, exogenous hormones could overcome seasonal constraints to produce competent oocytes with successful development in vitro but not in vivo, suggesting that differences in the oviductal environment negatively affected IVD embryo development in the anovulatory season (Palomino et al., 2020Palomino JM, Mastromonaco GF, Cervantes MP, Mapletoft RJ, Anzar M, Adams GP. Effect of season and superstimulatory treatment on in vivo and in vitro embryo production in wood bison (Bison bison athabascae). Reprod Domest Anim. 2020;55(1):54-63. http://doi.org/10.1111/rda.13580. PMid:31661568.
http://doi.org/10.1111/rda.13580...
). Subsequent attempts to produce embryos across ovulatory and anovulatory seasons focused on retrieving oocytes for IVP (Zwiefelhofer et al., 2022aZwiefelhofer ML, Singh J, Zwiefelhofer EM, Mastromonaco GF, Adams GP. Influence of ovarian follicular wave synchronization and single-dose eCG superstimulation on oocyte collection and in vitro embryo production in bison during the ovulatory and anovulatory seasons. Theriogenology. 2022a;187:238-46. http://doi.org/10.1016/j.theriogenology.2022.04.023. PMid:35660373.
http://doi.org/10.1016/j.theriogenology....
). To date, >20 calves have been born from AI, MOET and IVPET using fresh-chilled, frozen-thawed and sex-sorted sperm (reviewed by Acevedo and Barfield, 2023Acevedo C, Barfield JP. Review: reproductive physiology of bison and application of assisted reproductive technologies to their conservation. Animal. 2023;17(Suppl 1):100842. http://doi.org/10.1016/j.animal.2023.100842. PMid:37567664.
http://doi.org/10.1016/j.animal.2023.100...
). With progress now being made on the collection of sperm and oocytes from free-ranging wood bison (Zwiefelhofer et al., 2022bZwiefelhofer ML, Shury T, Zwiefelhofer EM, Singh J, Mastromonaco GF, Adams GP. Strategies for oocyte collection and in vitro embryo production in free-roaming bison herds. Conserv Physiol. 2022b;10(1):coac058. http://doi.org/10.1093/conphys/coac058. PMid: 35966755.
http://doi.org/10.1093/conphys/coac058...
), the wood bison project is one of the applications of the One Plan Approach to conservation that will include biobanking and ARTs; a potential model for the wisent, another bison species at risk (Duszewska et al., 2022Duszewska AM, Baraniewicz-Kołek M, Wojdan J, Barłowska K, Bielecki W, Gręda P, Niżański W, Olech W. Establishment of a wisent (Bison bonasus) germplasm bank. Animals (Basel). 2022;12(10):1239. http://doi.org/10.3390/ani12101239. PMid:35625085.
http://doi.org/10.3390/ani12101239...
).

c) In Vitro Produced Embryo Transfer (IVPET)

IVPET is the goal of many wildlife ART programs interested in preserving female genetics. Both sperm and oocytes are taken outside their ‘natural habitats’ and moved ex vivo into the culture dish requiring in-depth knowledge of gamete maturation, fertilization, and embryo development to ensure post-implantation success. Studies have highlighted the differences between IVP and IVD embryos, with the latter typically resulting in greater success post-thaw, -implantation and -birth (Ferré et al., 2020Ferré LB, Kjelland ME, Strøbech LB, Hyttel P, Mermillod P, Ross PJ. Review: recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods. Animal. 2020;14(5):991-1004. http://doi.org/10.1017/S1751731119002775. PMid:31760966.
http://doi.org/10.1017/S1751731119002775...
). While domestic and laboratory animal models have been instrumental in generating basic protocols, species-specific requirements have made advancement challenging in many wildlife species. Aquatic external fertilizers (fishes and amphibians), which release large numbers of oocytes and do not require the final embryo transfer step to produce live young, have experienced the greatest successes with IVP embryos. Aside from these cases, embryo technologies are not currently being implemented in species restoration programs. Wildlife IVP embryos have been created by both conventional (IVF) and advanced (somatic cell nuclear transfer; SCNT) methods to begin populating the conservation biobanks with female genetics (Mastromonaco, 2024Mastromonaco GF. A quarter century of CANDES: the state of embryo technologies in companion animals, non-domestic and endangered species. Theriogenology Wild. 2024;4:100069. http://doi.org/10.1016/j.therwi.2023.100069.
http://doi.org/10.1016/j.therwi.2023.100...
). Optimistically, in vitro gametogenesis (IVG) may be contributing to the growing list of banked gametes and embryos in the coming years. To date, animals born from IVPET have been scarce such that announcements of confirmed pregnancies or births continue to generate media attention.

  1. In Vitro Fertilization (IVF)

Challenges with in vitro maturation, fertilization, and culture (IVM-IVF-IVC), typically reported as low polar body extrusion, cleavage and blastocyst rates in domestic species, are also experienced in many wildlife species (Mastromonaco, 2024Mastromonaco GF. A quarter century of CANDES: the state of embryo technologies in companion animals, non-domestic and endangered species. Theriogenology Wild. 2024;4:100069. http://doi.org/10.1016/j.therwi.2023.100069.
http://doi.org/10.1016/j.therwi.2023.100...
). For instance, ovulation of immature (MI) oocytes and difficulties with in vitro maturation (<20% MII) in domestic dogs results in the need for in vivo matured oocytes in gray wolf (Canis lupus) IVPET trials (Nagashima and Songsasen, 2021Nagashima JB, Songsasen N. Canid reproductive biology: norm and unique aspects in strategies and mechanisms. Animals (Basel). 2021;11(3):653. http://doi.org/10.3390/ani11030653. PMid:33804569.
http://doi.org/10.3390/ani11030653...
). Similarly, a lack of optimal IVM systems for domestic cats (~60% MII) drives the outcomes in wild felid studies (Thongphakdee et al., 2020Thongphakdee A, Sukparangsi W, Comizzoli P, Chatdarong K. Reproductive biology and biotechnologies in wild felids. Theriogenology. 2020;150:360-73. http://doi.org/10.1016/j.theriogenology.2020.02.004. PMid:32102745.
http://doi.org/10.1016/j.theriogenology....
), and poor in vitro capacitation in domestic horse sperm leads to the use of intracytoplasmic sperm injection (ICSI) in zebra, Przewalski’s horse (Gambini et al., 2020Gambini A, Duque Rodríguez M, Rodríguez MB, Briski O, Flores Bragulat AP, Demergassi N, Losinno L, Salamone DF. Horse ooplasm supports in vitro preimplantation development of zebra ICSI and SCNT embryos without compromising YAP1 and SOX2 expression pattern. PLoS One. 2020;15(9):e0238948. http://doi.org/10.1371/journal.pone.0238948. PMid:32915925.
http://doi.org/10.1371/journal.pone.0238...
), and white rhinoceros (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...
). Inadequate IVC conditions responsible for the developmental ‘block’ at the time of embryonic genome activation in novel species has required transfer of embryos in the early cleavage stages in springbok and blesbok (Antidorcas marsupialis and Damaliscus pygargus, respectively; Chatiza et al., 2013Chatiza FP, Bartels P, Nedambale TL, Wagenaar GM. Sperm-egg interaction and functional assessment of springbok, impala and blesbok cauda epididymal spermatozoa using a domestic cattle in vitro fertilization system. Anim Reprod Sci. 2013;143(1-4):8-18. http://doi.org/10.1016/j.anireprosci.2013.11.001. PMid:24284137.
http://doi.org/10.1016/j.anireprosci.201...
), Eld’s deer (Rucervus eldii; Thongphakdee et al., 2017Thongphakdee A, Berg DK, Tharasanit T, Thongtip N, Tipkantha W, Punkong C, Tongthainan D, Noimoon S, Maikeaw U, Kajornklin N, Siriaroonrat B, Comizzoli P, Kamolnorranath S. The impact of ovarian stimulation protocol on oocyte quality, subsequent in vitro embryo development, and pregnancy after transfer to recipients in Eld’s deer (Rucervus eldii thamin). Theriogenology. 2017;91:134-44. http://doi.org/10.1016/j.theriogenology.2016.12.021. PMid:28215678.
http://doi.org/10.1016/j.theriogenology....
), and reindeer (Rangifer tarandus; Lindeberg et al., 2021Lindeberg H, Nikitkina E, Nagy S, Musidray A, Shiryaev G, Kumpula J, Holand Ø. Potential applications of assisted reproductive technologies (ART) in reindeer (Rangifer tarandus). Anim Reprod Sci. 2021;235:106890. http://doi.org/10.1016/j.anireprosci.2021.106890. PMid:34847527.
http://doi.org/10.1016/j.anireprosci.202...
). In non-mammalian species, yolk-laden oocytes have made cryopreservation significantly more challenging than the smaller mammalian oocytes (Diwan et al., 2020Diwan AD, Harke SN, Gopalkrishna, Panche AN. Gopalkrishna, Panche AN. Cryobanking of fish and shellfish egg, embryos and larvae: an overview. Front Mar Sci. 2020;7:251. http://doi.org/10.3389/fmars.2020.00251.
http://doi.org/10.3389/fmars.2020.00251...
). Despite these setbacks, researchers have not only generated IVF embryos but produced offspring of genetic value to the captive and wild populations.

Spotlight: Brown Brocket Deer (Subulo gouazoubira)

Over the past decade, the work of José Duarte on brown brocket deer highlights the challenges in developing IVF programs for novel species. Using a multi-disciplinary approach to neotropical deer genetic preservation involving cytogenetics, molecular genetics, and reproductive biology, Duarte and colleagues have been investigating the full spectrum of ARTs (Rola et al., 2021aRola LD, Buzanskas ME, Melo LM, Chaves MS, Freitas VJF, Duarte JMB. Assisted reproductive technology in neotropical deer: a model approach to preserving genetic diversity. Animals (Basel). 2021a;11(7):1961. http://doi.org/10.3390/ani11071961. PMid:34209061.
http://doi.org/10.3390/ani11071961...
). Common brown brocket deer pregnancies were obtained using frozen-thawed sperm, potentially serving as a model for more vulnerable brocket deer species (Duarte et al., 2023Duarte GS, Galindo DJ, Baldini MHM, da Fonseca JF, Duarte JMB, Oliveira MEF. Transcervical artificial insemination in the brown brocket deer (Subulo gouazoubira): a promising method for assisted reproduction in deer. Sci Rep. 2023;13(1):17369. http://doi.org/10.1038/s41598-023-43392-4. PMid:37833322.
http://doi.org/10.1038/s41598-023-43392-...
). Retrieval of oocytes from super-stimulated ovaries using laparascopic techniques resulted in 65% IVM rates (Rola et al., 2021bRola LD, Zanetti EDS, Del Collado M, Peroni EFC, Duarte JMB. Collection and in vitro maturation of Mazama gouazoubira (brown brocket deer) oocytes obtained after ovarian stimulation. Zygote. 2021b;29(3):216-22. http://doi.org/10.1017/S0967199420000787. PMid:33446301.
http://doi.org/10.1017/S0967199420000787...
). While embryos have not yet been produced in brown brocket deer by conventional IVF methods, iSCNT (using domestic cattle oocytes) resulted in 6% blastocysts (Melo et al., 2022Melo LM, Silva SB, Magalhães LC, Cortez JV, Kumar S, Duarte JMB, Rola LD, Chaves MS, Freitas VJF. The use of somatic cell nuclear transfer to obtain interspecific cloned embryos from brown brocket deer karyoplast and bovine cytoplast: embryo development and nuclear gene expression. Theriogenology Wild. 2022;1:100001. http://doi.org/10.1016/j.therwi.2022.100001.
http://doi.org/10.1016/j.therwi.2022.100...
). Notably, progress in wild cervid IVF has been challenging with low blastocyst rates observed in Eld’s deer (5%; Thongphakdee et al., 2017Thongphakdee A, Berg DK, Tharasanit T, Thongtip N, Tipkantha W, Punkong C, Tongthainan D, Noimoon S, Maikeaw U, Kajornklin N, Siriaroonrat B, Comizzoli P, Kamolnorranath S. The impact of ovarian stimulation protocol on oocyte quality, subsequent in vitro embryo development, and pregnancy after transfer to recipients in Eld’s deer (Rucervus eldii thamin). Theriogenology. 2017;91:134-44. http://doi.org/10.1016/j.theriogenology.2016.12.021. PMid:28215678.
http://doi.org/10.1016/j.theriogenology....
), and reindeer (4%; Peippo et al., 2019Peippo J, Lindeberg H, Mutikainen M, Harmama T-M, Kumpula J, Holand O. In vitro production of the first reindeer (Rangifer tarandus tarandus) blastocysts. In: Proceedings of the 35th annual meeting of the European Embryo Transfer Association; 2019 Sep 12-14; Murcia, Spain. Murcia: AETE; 2019. p. 746.). In contrast, dedicated efforts in the Japanese sika deer (Cervus nippon nippon), a model for the endangered Vietnamese sika deer (Cervus nippon pseudaxis), resulted in 30% blastocyst rates once the embryos were co-cultured with ovine epithelial cells (Locatelli et al., 2012Locatelli Y, Hendriks A, Vallet JC, Baril G, Duffard N, Bon N, Ortiz K, Scala C, Maurel MC, Mermillod P, Legendre X. Assessment LOPU-IVF in Japanese sika deer (Cervus nippon nippon) and application to Vietnamese sika deer (Cervus nippon pseudaxis) a related subspecies threatened with extinction. Theriogenology. 2012;78(9):2039-49. http://doi.org/10.1016/j.theriogenology.2012.07.025. PMid:23043947.
http://doi.org/10.1016/j.theriogenology....
). With so many species in need of assistance, prioritizing resources based on ecosystem or food security relevance will be essential for achieving conservation impact with ARTs.

  1. Somatic Cell Nuclear Transfer (SCNT)

Following the report by Dominko et al (1999)Dominko T, Mitalipova M, Haley B, Beyhan Z, Memili E, McKusick B, First NL. Bovine oocyte cytoplasm supports development of embryos produced by nuclear transfer of somatic cell nuclei from various mammalian species. Biol Reprod. 1999;60(6):1496-502. http://doi.org/10.1095/biolreprod60.6.1496. PMid:10330111.
http://doi.org/10.1095/biolreprod60.6.14...
demonstrating the bovine ooplasm’s capacity as a ‘universal recipient’, interspecies SCNT (iSCNT) emerged, fueling the rise in somatic cell biobanks in zoological and government institutions, as well as the production of iSCNT embryos in more than 50 species (Mastromonaco et al., 2014Mastromonaco GF, González-Grajales LA, Filice M, Comizzoli P. Somatic cells, stem cells, and induced pluripotent stem cells: how do they now contribute to conservation? In: Holt WV, Brown JL, Comizzoli P, editors. Reproductive sciences in animal conservation. advances in experimental medicine and biology. New York: Springer; 2014. p. 385-427. http://doi.org/10.1007/978-1-4939-0820-2_16.
http://doi.org/10.1007/978-1-4939-0820-2...
). Although blastocyst development has been demonstrated in diverse species, including tiger (Panthera tigris), red panda (Ailurus fulgens), and minke whale (Balaenoptera acutorostrata), only a small number of offspring have been born (Mastromonaco et al., 2014Mastromonaco GF, González-Grajales LA, Filice M, Comizzoli P. Somatic cells, stem cells, and induced pluripotent stem cells: how do they now contribute to conservation? In: Holt WV, Brown JL, Comizzoli P, editors. Reproductive sciences in animal conservation. advances in experimental medicine and biology. New York: Springer; 2014. p. 385-427. http://doi.org/10.1007/978-1-4939-0820-2_16.
http://doi.org/10.1007/978-1-4939-0820-2...
). Challenges stemming from low and aberrant reprogramming have impacted the advancement of iSCNT as a practical tool for offspring production. From a species conservation perspective, the recent birth of the black footed ferret clone using donor cells cryopreserved from a wild female in 1988 and oocytes from domestic ferrets provided an opportunity to re-introduce under-represented genetics, thereby having a significant impact on the diversity of the remaining ferret population (Imbler, 2021Imbler S. [homepage on the Internet]. USA: The New York TImes; 2021. Meet Elizabeth Ann, the first cloned black-footed ferret; 2021 [cited 2024 February 18]. Available from: https://www.nytimes.com/2021/02/18/science/black-footed-ferret-clone.html.
https://www.nytimes.com/2021/02/18/scien...
). This is a key example of the potential for cloning technologies in the One Plan Approach as it was always known that iSCNT would not become a widespread tool for offspring production, but a powerful tool for the ‘resurrection’ of key individuals of high genetic value.

Spotlight: Przewalski’s Horse (Equus ferus przewalskii)

Species with small founder gene pools and high inbreeding coefficients typically require reproductive support as with the Przewalski’s horse. Although the first retrievals and interspecies transfers of Przewalski’s horse embryos occurred more than 40 years ago (Allen and Wilsher, 2020Allen WRT, Wilsher S. Historical aspects of equine embryo transfer. J Equine Vet Sci. 2020;89:102987. http://doi.org/10.1016/j.jevs.2020.102987. PMid:32563443.
http://doi.org/10.1016/j.jevs.2020.10298...
), there has been only one reported birth of a healthy foal following timed AI and none from IVPET reviewed by Cabeza and Gambini, 2023Cabeza JP, Gambini A. Advancements and challenges in in vitro reproductive technologies for the conservation of equine species. Theriogenology Wild. 2023;2:100036. http://doi.org/10.1016/j.therwi.2023.100036.
http://doi.org/10.1016/j.therwi.2023.100...
). Advancements in domestic horse SCNT (approximately 400 foals born; reviewed by Gambini and Maserati, 2017Gambini A, Maserati M. A journey through horse cloning. Reprod Fertil Dev. 2017;30(1):8-17. http://doi.org/10.1071/RD17374. PMid:29539299.
http://doi.org/10.1071/RD17374...
) fostered the opportunity to investigate iSCNT as a tool to produce Przewalski’s horse foals. Under the guidance of Oliver Ryder with the San Diego Zoo’s Frozen Zoo®, Przewalski’s horse stallion cells cryopreserved in 1980 were used to produce 11 transferrable embryos that resulted in 7 pregnancies and 2 live births (Novak et al., 2024Novak BJ, Ryder OA, Houck ML, Putnam AS, Walker K, Russell L, Russell B, Walker S, Arenivas SS, Aston L, Veneklasen G, Ivy JA, Koepfli K-P, Rusnak A, Simek J, Zhuk A, Phelan R. Endangered Przewalski’s horse, Equus przewalskii, cloned from historically cryopreserved cells. bioRxiv 2024;1-17. http://doi.org/10.1101/2023.12.20.572538.
http://doi.org/10.1101/2023.12.20.572538...
). While it may not be a significant scientific advancement, it is a remarkable change in mind-set as the American Association of Zoos and Aquariums previously did not accept nuclear-cytoplasmic hybrids into the captive gene pool. Further, the project was supported by the not-for-profit Revive & Restore (2024), aRevive & Restore. [homepage on the internet]. Revive & Restore; 2024. The genetic rescue toolkit; 2024 [cited 2024 March 10]. Available from: https://reviverestore.org/what-we-do/genetic-rescue-toolkit
https://reviverestore.org/what-we-do/gen...
sponsor of de-extinction projects such as the woolly mammoth, passenger pigeon, great auk and more (Revive & Restore, 2024Revive & Restore. [homepage on the internet]. Revive & Restore; 2024. The genetic rescue toolkit; 2024 [cited 2024 March 10]. Available from: https://reviverestore.org/what-we-do/genetic-rescue-toolkit
https://reviverestore.org/what-we-do/gen...
), which may be academically interesting pursuits but do not serve the conservation community.

  1. In Vitro Gametogenesis (IVG)

The potential to ‘convert’ adult somatic cells into induced pluripotent stem cells (iPSCs) using human and mouse transcription factors (‘Yamanaka factors’; Ogorevc et al., 2016Ogorevc J, Orehek S, Dovč P. Cellular reprogramming in farm animals: an overview of iPSC generation in the mammalian farm animal species. J Anim Sci Biotechnol. 2016;7(1):10. http://doi.org/10.1186/s40104-016-0070-3. PMid:26900466.
http://doi.org/10.1186/s40104-016-0070-3...
) captivated researchers interested in understanding the cellular and developmental biology of wildlife species. Since then, iPSCs have been generated in >20 wildlife species, including snow leopard (Panthera uncia), Sumatran orangutan (Pongo abelii), little brown bat (Myotis lucifugus), Okinawa rail (Hypotaenidia okinawae), Japanese ptarmigan (Lagopus muta japonica), to name a few (Katayama et al., 2022Katayama M, Fukuda T, Kaneko T, Nakagawa Y, Tajima A, Naito M, Ohmaki H, Endo D, Asano M, Nagamine T, Nakaya Y, Saito K, Watanabe Y, Tani T, Inoue-Murayama M, Nakajima N, Onuma M. Induced pluripotent stem cells of endangered avian species. Commun Biol. 2022;5(1):1049. http://doi.org/10.1038/s42003-022-03964-y. PMid:36280684.
http://doi.org/10.1038/s42003-022-03964-...
; Swegen et al., 2023Swegen A, Appeltant R, Williams SA. Cloning in action: can embryo splitting, induced pluripotency and somatic cell nuclear transfer contribute to endangered species conservation? Biol Rev Camb Philos Soc. 2023;98(4):1225-49. http://doi.org/10.1111/brv.12951. PMid:37016502.
http://doi.org/10.1111/brv.12951...
). Advancements in stem cell induction and differentiation brought IVG closer to reality with the birth of mouse pups following in vitro oogenesis that generated the iPSC-derived oocytes (Hayashi and Saitou, 2013Hayashi K, Saitou M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protoc. 2013;8(8):1513-24. http://doi.org/10.1038/nprot.2013.090. PMid:23845963.
http://doi.org/10.1038/nprot.2013.090...
). More recently, in vitro spermatogenesis resulted in iPSC-derived spermatids used to produce living offspring in the rat (Matsumura et al., 2023Matsumura T, Katagiri K, Yao T, Ishikawa-Yamauchi Y, Nagata S, Hashimoto K, Sato T, Kimura H, Shinohara T, Sanbo M, Hirabayashi M, Ogawa T. Generation of rat offspring using spermatids produced through in vitro spermatogenesis. Sci Rep. 2023;13(1):12105. http://doi.org/10.1038/s41598-023-39304-1. PMid:37495678.
http://doi.org/10.1038/s41598-023-39304-...
). Similar to SCNT, iPSC-based IVG requires successful reprogramming of adult somatic cells into a stable pluripotent state, a feat that is not easily achieved (Li et al., 2014Li J, Song W, Pan G, Zhou J. Advances in understanding the cell types and approaches used for generating induced pluripotent stem cells. J Hematol Oncol. 2014;7(1):50. http://doi.org/10.1186/s13045-014-0050-z. PMid:25037625.
http://doi.org/10.1186/s13045-014-0050-z...
). Further, differentiation of the iPSCs into primordial germ cell-like cells (PGCLCs) has been effective in the mouse model but difficult to achieve in farm animals (Strange and Alberio, 2023Strange A, Alberio R. Review: a barnyard in the lab: prospect of generating animal germ cells for breeding and conservation. Animal. 2023;17(Suppl 1):100753. http://doi.org/10.1016/j.animal.2023.100753. PMid:37567650.
http://doi.org/10.1016/j.animal.2023.100...
). This comes with additional safety challenges due to the teratogenic potential that arises from using transgene integrating delivery methods and a highly oncogenic set of genes. There is hope that the significant efforts already underway in both the human and animal fields will propel IVG science forward.

Spotlight: White Rhinoceros (Ceratotherium simum)

Since the 1990s, cases of ovarian cycle irregularities and reproductive pathologies have been investigated in captive rhinoceros (reviewed by Roth, 2024Roth TL. That was then, this is now – over two decades of progress in rhinoceros reproductive science and technology. Theriogenology Wild. 2024;4:100065. http://doi.org/10.1016/j.therwi.2023.100065.
http://doi.org/10.1016/j.therwi.2023.100...
). However, the plight of the northern white rhinoceros (rhino), a functionally extinct subspecies with only two living adult females, instigated one of the most lucrative partnerships in conservation ART history between BioRescue and Colossal Biosciences (BusinessWire, 2023BusinessWire. [homepage on the internet]. BioSpace, 2023. Colossal biosciences joins BioRescue in its mission to save the northern white rhino from extinction; 2023 [cited 2024 March 20]. Available from: https://www.biospace.com/article/releases/colossal-biosciences-joins-biorescue-in-its-mission-to-save-the-northern-white-rhino-from-extinction/.
https://www.biospace.com/article/release...
). What began as technique development to support white rhino breeding programs has resulted in a race to de-extinct the northern white rhino subspecies. Multiple attempts at live and post-mortem oocyte collection followed by IVM-IVF-IVC using domestic horse protocols have recently resulted in blastocysts (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...
). While there have been 10 white rhino calves produced by AI with fresh and frozen sperm (Roth, 2024Roth TL. That was then, this is now – over two decades of progress in rhinoceros reproductive science and technology. Theriogenology Wild. 2024;4:100065. http://doi.org/10.1016/j.therwi.2023.100065.
http://doi.org/10.1016/j.therwi.2023.100...
), there are no live calves from the transfer of IVF embryos to date. Currently, there are two research teams that have independently induced a pluripotency state in multiple northern white rhino somatic cell lines, some of which had been cryopreserved for 40 years (Korody et al., 2021Korody ML, Ford SM, Nguyen TD, Pivaroff CG, Valiente-Alandi I, Peterson SE, Ryder OA, Loring JF. Rewinding extinction in the northern white rhinoceros: genetically diverse induced pluripotent stem cell bank for genetic rescue. Stem Cells Dev. 2021;30(4):177-89. http://doi.org/10.1089/scd.2021.0001. PMid:33406994.
http://doi.org/10.1089/scd.2021.0001...
; 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-...
). Progress towards IVG was reported in 2022 with the generation of primordial germ cell-like cells northern white rhino iPSCs (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...
). While these initiatives don’t have immediate application in biodiversity conservation, they are important advancements towards the establishment of somatic cell technologies for genetic management of wildlife species.

Focus for the Future

Forty years of efforts from researchers around the world have resulted in a proportionately small number of cases that have successfully integrated AI, MOET or IVPET into species conservation plans. To date, ARTs have been more readily applied in external fertilizers, specifically fishes and amphibians, since the in vitro environment does not have the complexity of re-creating the oviductal or uterine milieu ex vivo. In contrast, mammals will not experience the same widespread application and abundance in offspring production anytime soon. There are several key research priorities to continue advancing mammalian ARTs: a) the oocyte (culture optimization and cryopreservation), b) the pregnant uterus (recipient management), and c) the environment (emerging pollutants and climate change). Innovative technologies, ranging from microfluidics to 3-D culture systems (Ferraz and Ferronato, 2023Ferraz MAMM, Ferronato GA. Opportunities involving microfluidics and 3D culture systems to the in vitro embryo production. Anim Reprod. 2023;20(2):e20230058. http://doi.org/10.1590/1984-3143-ar2023-0058. PMid:37638255.
http://doi.org/10.1590/1984-3143-ar2023-...
), offer untapped possibilities for overcoming the current limitations in producing embryos with optimal developmental capacity pre- and post-implantation. Similarly, the growing literature on nutrition provides important insights for improving pregnancy maintenance and fetal development (Wyse et al., 2022Wyse J, Latif S, Gurusinghe S, McCormick J, Weston LA, Stephen CP. Phytoestrogens: a review of their impacts on reproductive physiology and other effects upon grazing livestock. Animals (Basel). 2022;12(19):2709. http://doi.org/10.3390/ani12192709. PMid:36230450.
http://doi.org/10.3390/ani12192709...
). Unfortunately, reproductive health and ART success will be challenged by the continuously changing natural environments, particularly ubiquitous particles such as micro- and nano-plastics (Aardema et al., 2024Aardema H, Vethaak A, Kamstra JH, Legler J. Farm animals as a critical link between environmental and human health impacts of micro-and nanoplastics. Micropl and Nanopl. 2024;4(5):5. http://doi.org/10.1186/s43591-024-00082-w.
http://doi.org/10.1186/s43591-024-00082-...
).

As species continue to face threats to their long-term survival, ARTs provide some assurance that the genetics carefully stewarded in wildlife biobanks around the globe can be used to keep keystone species thriving and ecosystems intact. For ARTs to become a realistic addition to the One Plan Approach to species conservation, comprehensive programs must include not only the initial research and protocol development phase, but also establishment of partnerships and generation of banked inventory through systematic sample collection. In a field with such limited resources, it is important to remain focused on species or populations that have the greatest chance for self-sustainability in the long term. Thus, projects directing resources and attention towards extinct or functionally extinct species, while making potentially valuable scientific advancements that could be extrapolated to related species, are a distraction from impact-driven species conservation objectives.

Acknowledgements

Thanks to Cristina Lyons for assistance with reference compilation for the manuscript and Virginia Wolfe for preparation of Figure 1. Special thanks to Dr. Ariel Kahrl for providing us with the original files of the sperm images used in Figure 1.

  • Financial support: None.
  • How to cite: Mastromonaco G. 40 ‘wild’ years: the current reality and future potential of assisted reproductive technologies in wildlife species. Anim Reprod. 2024;21(3):e20240049. https://doi.org/10.1590/1984-3143-AR2024-0049

References

  • Aardema H, Vethaak A, Kamstra JH, Legler J. Farm animals as a critical link between environmental and human health impacts of micro-and nanoplastics. Micropl and Nanopl. 2024;4(5):5. http://doi.org/10.1186/s43591-024-00082-w
    » http://doi.org/10.1186/s43591-024-00082-w
  • Acevedo C, Barfield JP. Review: reproductive physiology of bison and application of assisted reproductive technologies to their conservation. Animal. 2023;17(Suppl 1):100842. http://doi.org/10.1016/j.animal.2023.100842 PMid:37567664.
    » http://doi.org/10.1016/j.animal.2023.100842
  • Allen WRT, Wilsher S. Historical aspects of equine embryo transfer. J Equine Vet Sci. 2020;89:102987. http://doi.org/10.1016/j.jevs.2020.102987 PMid:32563443.
    » http://doi.org/10.1016/j.jevs.2020.102987
  • Anzalone DA, Palazzese L, Iuso D, Martino G, Loi P. Freeze-dried spermatozoa: an alternative biobanking option for endangered species. Anim Reprod Sci. 2018;190:85-93. http://doi.org/10.1016/j.anireprosci.2018.01.010 PMid:29397252.
    » http://doi.org/10.1016/j.anireprosci.2018.01.010
  • Baldrighi JM, Sá Filho MF, Siqueira AF, Visintin JA, Baruselli PS, Assumpção MEOA. Temporal evaluation of follicular dynamics and endocrine patterns of Holstein (Bos taurus), Gir (Bos indicus), and Murrah (Bubalus bubalis) heifers kept under the same nutritional, management and environmental conditions. Theriogenology. 2022;190:8-14. http://doi.org/10.1016/j.theriogenology.2022.07.006 PMid:35863098.
    » http://doi.org/10.1016/j.theriogenology.2022.07.006
  • Ballou JD. Strategies for maintaining genetic diversity in captive populations through reproductive technology. Zoo Biol. 1984;3(4):311-23. http://doi.org/10.1002/zoo.1430030404
    » http://doi.org/10.1002/zoo.1430030404
  • Beirão J, Boulais M, Gallego V, O’Brien JK, Peixoto S, Robeck TR, Cabrita E. Sperm handling in aquatic animals for artificial reproduction. Theriogenology. 2019;133:161-78. http://doi.org/10.1016/j.theriogenology.2019.05.004 PMid:31108371.
    » http://doi.org/10.1016/j.theriogenology.2019.05.004
  • Bison Integrated Genomics (BIG) Project. [homepage on the Internet]. Parks Canada; 2022 [cited 2024 March 9]. Available from: https://www.bigproject.ca/
    » https://www.bigproject.ca/
  • Blanco JM, Wildt DE, Höfle U, Voelker W, Donoghue AM. Implementing artificial insemination as an effective tool for ex situ conservation of endangered avian species. Theriogenology. 2009;71(1):200-13. http://doi.org/10.1016/j.theriogenology.2008.09.019 PMid:19004491.
    » http://doi.org/10.1016/j.theriogenology.2008.09.019
  • BusinessWire. [homepage on the internet]. BioSpace, 2023. Colossal biosciences joins BioRescue in its mission to save the northern white rhino from extinction; 2023 [cited 2024 March 20]. Available from: https://www.biospace.com/article/releases/colossal-biosciences-joins-biorescue-in-its-mission-to-save-the-northern-white-rhino-from-extinction/
    » https://www.biospace.com/article/releases/colossal-biosciences-joins-biorescue-in-its-mission-to-save-the-northern-white-rhino-from-extinction/
  • Byers O, Lees C, Wilcken J, Schwitzer C. The One Plan Approach: the philosophy and implementation of CBSG’s approach to integrated species conservation planning. In: Gusset M, Dick G, editors. Towards integrated species conservation. Vol. 14. Switzerland: World Association of Zoos and Aquariums Magazine. [serial on the internet]. 2013 [cited 2024 March 9]. p. 2-5. Available from: https://www.waza.org/wp-content/uploads/2019/02/waza_mag_14.pdf
    » https://www.waza.org/wp-content/uploads/2019/02/waza_mag_14.pdf
  • Cabeza JP, Gambini A. Advancements and challenges in in vitro reproductive technologies for the conservation of equine species. Theriogenology Wild. 2023;2:100036. http://doi.org/10.1016/j.therwi.2023.100036
    » http://doi.org/10.1016/j.therwi.2023.100036
  • Chatiza FP, Bartels P, Nedambale TL, Wagenaar GM. Sperm-egg interaction and functional assessment of springbok, impala and blesbok cauda epididymal spermatozoa using a domestic cattle in vitro fertilization system. Anim Reprod Sci. 2013;143(1-4):8-18. http://doi.org/10.1016/j.anireprosci.2013.11.001 PMid:24284137.
    » http://doi.org/10.1016/j.anireprosci.2013.11.001
  • Crosier AE, Comizzoli P, Koester DC, Wildt DE. Circumventing the natural, frequent oestrogen waves of the female cheetah (Acinonyx jubatus) using oral progestin (Altrenogest). Reprod Fertil Dev. 2017;29(8):1486-98. http://doi.org/10.1071/RD16007 PMid:27483379.
    » http://doi.org/10.1071/RD16007
  • Crosier AE, Henghali JN, Howard J, Pukazhenthi BS, Terrell KA, Marker LL, Wildt DE. Improved quality of cryopreserved cheetah (Acinonyx jubatus) spermatozoa after centrifugation through Accudenz. J Androl. 2009;30(3):298-308. http://doi.org/10.2164/jandrol.108.006239 PMid:19023140.
    » http://doi.org/10.2164/jandrol.108.006239
  • Diwan AD, Harke SN, Gopalkrishna, Panche AN. Gopalkrishna, Panche AN. Cryobanking of fish and shellfish egg, embryos and larvae: an overview. Front Mar Sci. 2020;7:251. http://doi.org/10.3389/fmars.2020.00251
    » http://doi.org/10.3389/fmars.2020.00251
  • Dominko T, Mitalipova M, Haley B, Beyhan Z, Memili E, McKusick B, First NL. Bovine oocyte cytoplasm supports development of embryos produced by nuclear transfer of somatic cell nuclei from various mammalian species. Biol Reprod. 1999;60(6):1496-502. http://doi.org/10.1095/biolreprod60.6.1496 PMid:10330111.
    » http://doi.org/10.1095/biolreprod60.6.1496
  • Dresser BL. Cryobiology, embryo transfer, and artificial insemination in ex situ animal conservation programs. In: Wilson EO, Peter FM, editors. Biodiversity. Washington (DC): National Academies Press; 1988. Chapter 34, p. 296-308.
  • Dresser BL. Embryo transfer in exotic bovids. Int Zoo Yearb. 1986;24(1):138-42. http://doi.org/10.1111/j.1748-1090.1985.tb02529.x
    » http://doi.org/10.1111/j.1748-1090.1985.tb02529.x
  • Duarte GS, Galindo DJ, Baldini MHM, da Fonseca JF, Duarte JMB, Oliveira MEF. Transcervical artificial insemination in the brown brocket deer (Subulo gouazoubira): a promising method for assisted reproduction in deer. Sci Rep. 2023;13(1):17369. http://doi.org/10.1038/s41598-023-43392-4 PMid:37833322.
    » http://doi.org/10.1038/s41598-023-43392-4
  • Duszewska AM, Baraniewicz-Kołek M, Wojdan J, Barłowska K, Bielecki W, Gręda P, Niżański W, Olech W. Establishment of a wisent (Bison bonasus) germplasm bank. Animals (Basel). 2022;12(10):1239. http://doi.org/10.3390/ani12101239 PMid:35625085.
    » http://doi.org/10.3390/ani12101239
  • Ferraz MAMM, Ferronato GA. Opportunities involving microfluidics and 3D culture systems to the in vitro embryo production. Anim Reprod. 2023;20(2):e20230058. http://doi.org/10.1590/1984-3143-ar2023-0058 PMid:37638255.
    » http://doi.org/10.1590/1984-3143-ar2023-0058
  • Ferraz MAMM, Nagashima JB, Noonan MJ, Crosier AE, Songsasen N. Oviductal extracellular vesicles improve post-thaw sperm function in red wolves and cheetahs. Int J Mol Sci. 2020;21(10):3733. http://doi.org/10.3390/ijms21103733 PMid:32466321.
    » http://doi.org/10.3390/ijms21103733
  • Ferré LB, Kjelland ME, Strøbech LB, Hyttel P, Mermillod P, Ross PJ. Review: recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods. Animal. 2020;14(5):991-1004. http://doi.org/10.1017/S1751731119002775 PMid:31760966.
    » http://doi.org/10.1017/S1751731119002775
  • Gambini A, Duque Rodríguez M, Rodríguez MB, Briski O, Flores Bragulat AP, Demergassi N, Losinno L, Salamone DF. Horse ooplasm supports in vitro preimplantation development of zebra ICSI and SCNT embryos without compromising YAP1 and SOX2 expression pattern. PLoS One. 2020;15(9):e0238948. http://doi.org/10.1371/journal.pone.0238948 PMid:32915925.
    » http://doi.org/10.1371/journal.pone.0238948
  • Gambini A, Maserati M. A journey through horse cloning. Reprod Fertil Dev. 2017;30(1):8-17. http://doi.org/10.1071/RD17374 PMid:29539299.
    » http://doi.org/10.1071/RD17374
  • Hayashi K, Saitou M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protoc. 2013;8(8):1513-24. http://doi.org/10.1038/nprot.2013.090 PMid:23845963.
    » http://doi.org/10.1038/nprot.2013.090
  • 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
  • Herrick JR. Assisted reproductive technologies for endangered species conservation: developing sophisticated protocols with limited access to animals with unique reproductive mechanisms. Biol Reprod. 2019;100(5):1158-70. http://doi.org/10.1093/biolre/ioz025 PMid:30770538.
    » http://doi.org/10.1093/biolre/ioz025
  • 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
  • Holt WV, Fazeli A. Sperm storage in the female reproductive tract. Annu Rev Anim Biosci. 2016;4(1):291-310. http://doi.org/10.1146/annurev-animal-021815-111350 PMid:26526545.
    » http://doi.org/10.1146/annurev-animal-021815-111350
  • Imbler S. [homepage on the Internet]. USA: The New York TImes; 2021. Meet Elizabeth Ann, the first cloned black-footed ferret; 2021 [cited 2024 February 18]. Available from: https://www.nytimes.com/2021/02/18/science/black-footed-ferret-clone.html
    » https://www.nytimes.com/2021/02/18/science/black-footed-ferret-clone.html
  • Kahrl AF, Snook RR, Fitzpatrick JL. Fertilization mode differentially impacts the evolution of vertebrate sperm components. Nat Commun. 2022;13(1):6809. http://doi.org/10.1038/s41467-022-34609-7 PMid:36357384.
    » http://doi.org/10.1038/s41467-022-34609-7
  • Kaneko T, Ito H, Sakamoto H, Onuma M, Inoue-Murayama M. Sperm preservation by freeze-drying for the conservation of wild animals. PLoS One. 2014;9(11):e113381. http://doi.org/10.1371/journal.pone.0113381 PMid:25409172.
    » http://doi.org/10.1371/journal.pone.0113381
  • Katayama M, Fukuda T, Kaneko T, Nakagawa Y, Tajima A, Naito M, Ohmaki H, Endo D, Asano M, Nagamine T, Nakaya Y, Saito K, Watanabe Y, Tani T, Inoue-Murayama M, Nakajima N, Onuma M. Induced pluripotent stem cells of endangered avian species. Commun Biol. 2022;5(1):1049. http://doi.org/10.1038/s42003-022-03964-y PMid:36280684.
    » http://doi.org/10.1038/s42003-022-03964-y
  • Kersey DC, Wildt DE, Brown JL, Snyder RJ, Huang Y, Monfort SL. Endocrine milieu of perioestrus in the giant panda (Ailuropoda melanoleuca), as determined by non-invasive hormone measures. Reprod Fertil Dev. 2010;22(6):901-12. http://doi.org/10.1071/RD09178 PMid:20591324.
    » http://doi.org/10.1071/RD09178
  • Korody ML, Ford SM, Nguyen TD, Pivaroff CG, Valiente-Alandi I, Peterson SE, Ryder OA, Loring JF. Rewinding extinction in the northern white rhinoceros: genetically diverse induced pluripotent stem cell bank for genetic rescue. Stem Cells Dev. 2021;30(4):177-89. http://doi.org/10.1089/scd.2021.0001 PMid:33406994.
    » http://doi.org/10.1089/scd.2021.0001
  • Kouba AJ, Vance CK, Willis EL. Artificial fertilization for amphibian conservation: current knowledge and future considerations. Theriogenology. 2009;71(1):214-27. http://doi.org/10.1016/j.theriogenology.2008.09.055 PMid:19026442.
    » http://doi.org/10.1016/j.theriogenology.2008.09.055
  • Kouba AJ, Vance CK. Applied reproductive technologies and genetic resource banking for amphibian conservation. Reprod Fertil Dev. 2009;21(6):719-37. http://doi.org/10.1071/RD09038 PMid:19567216.
    » http://doi.org/10.1071/RD09038
  • Kydd J, Boyle MS, Allen WR, Shephard A, Summer PM. Transfer of exotic equine embryos to domestic horses and donkeys. Equine Vet J. 1985;17(S3):80-3. http://doi.org/10.1111/j.2042-3306.1985.tb04601.x
    » http://doi.org/10.1111/j.2042-3306.1985.tb04601.x
  • Li J, Song W, Pan G, Zhou J. Advances in understanding the cell types and approaches used for generating induced pluripotent stem cells. J Hematol Oncol. 2014;7(1):50. http://doi.org/10.1186/s13045-014-0050-z PMid:25037625.
    » http://doi.org/10.1186/s13045-014-0050-z
  • Lindeberg H, Nikitkina E, Nagy S, Musidray A, Shiryaev G, Kumpula J, Holand Ø. Potential applications of assisted reproductive technologies (ART) in reindeer (Rangifer tarandus). Anim Reprod Sci. 2021;235:106890. http://doi.org/10.1016/j.anireprosci.2021.106890 PMid:34847527.
    » http://doi.org/10.1016/j.anireprosci.2021.106890
  • Locatelli Y, Hendriks A, Vallet JC, Baril G, Duffard N, Bon N, Ortiz K, Scala C, Maurel MC, Mermillod P, Legendre X. Assessment LOPU-IVF in Japanese sika deer (Cervus nippon nippon) and application to Vietnamese sika deer (Cervus nippon pseudaxis) a related subspecies threatened with extinction. Theriogenology. 2012;78(9):2039-49. http://doi.org/10.1016/j.theriogenology.2012.07.025 PMid:23043947.
    » http://doi.org/10.1016/j.theriogenology.2012.07.025
  • Lueders I, Luther I, Scheepers G, van der Horst G. Improved semen collection method for wild felids: urethral catheterization yields high sperm quality in African lions (Panthera leo). Theriogenology. 2012;78(3):696-701. http://doi.org/10.1016/j.theriogenology.2012.02.026 PMid:22538007.
    » http://doi.org/10.1016/j.theriogenology.2012.02.026
  • Mastromonaco GF, González-Grajales LA, Filice M, Comizzoli P. Somatic cells, stem cells, and induced pluripotent stem cells: how do they now contribute to conservation? In: Holt WV, Brown JL, Comizzoli P, editors. Reproductive sciences in animal conservation. advances in experimental medicine and biology. New York: Springer; 2014. p. 385-427. http://doi.org/10.1007/978-1-4939-0820-2_16
    » http://doi.org/10.1007/978-1-4939-0820-2_16
  • Mastromonaco GF, Songsasen N. Reproductive technologies for the conservation of wildlife and endangered species. In: Presicce GA, editor. Reproductive technologies in animals. Cambridge: Academic Press; 2020. Chapter 7, p. 99-117.
  • Mastromonaco GF. A quarter century of CANDES: the state of embryo technologies in companion animals, non-domestic and endangered species. Theriogenology Wild. 2024;4:100069. http://doi.org/10.1016/j.therwi.2023.100069
    » http://doi.org/10.1016/j.therwi.2023.100069
  • Matsumura T, Katagiri K, Yao T, Ishikawa-Yamauchi Y, Nagata S, Hashimoto K, Sato T, Kimura H, Shinohara T, Sanbo M, Hirabayashi M, Ogawa T. Generation of rat offspring using spermatids produced through in vitro spermatogenesis. Sci Rep. 2023;13(1):12105. http://doi.org/10.1038/s41598-023-39304-1 PMid:37495678.
    » http://doi.org/10.1038/s41598-023-39304-1
  • Melo LM, Silva SB, Magalhães LC, Cortez JV, Kumar S, Duarte JMB, Rola LD, Chaves MS, Freitas VJF. The use of somatic cell nuclear transfer to obtain interspecific cloned embryos from brown brocket deer karyoplast and bovine cytoplast: embryo development and nuclear gene expression. Theriogenology Wild. 2022;1:100001. http://doi.org/10.1016/j.therwi.2022.100001
    » http://doi.org/10.1016/j.therwi.2022.100001
  • Menkhorst E, Selwood L. Vertebrate extracellular preovulatory and postovulatory egg coats. Biol Reprod. 2008;79(5):790-7. http://doi.org/10.1095/biolreprod.108.068551 PMid:18614703.
    » http://doi.org/10.1095/biolreprod.108.068551
  • Nagashima JB, Songsasen N. Canid reproductive biology: norm and unique aspects in strategies and mechanisms. Animals (Basel). 2021;11(3):653. http://doi.org/10.3390/ani11030653 PMid:33804569.
    » http://doi.org/10.3390/ani11030653
  • Novak BJ, Ryder OA, Houck ML, Putnam AS, Walker K, Russell L, Russell B, Walker S, Arenivas SS, Aston L, Veneklasen G, Ivy JA, Koepfli K-P, Rusnak A, Simek J, Zhuk A, Phelan R. Endangered Przewalski’s horse, Equus przewalskii, cloned from historically cryopreserved cells. bioRxiv 2024;1-17. http://doi.org/10.1101/2023.12.20.572538
    » http://doi.org/10.1101/2023.12.20.572538
  • O’Brien JK, Robeck TR. Development of sperm sexing and associated assisted reproductive technology for sex preselection of captive bottlenose dolphins (Tursiops truncatus). Reprod Fertil Dev. 2006;18(3):319-29. http://doi.org/10.1071/RD05108 PMid:16554007.
    » http://doi.org/10.1071/RD05108
  • O’Brien JK, Steinman KJ, Schmitt T, Robeck TR. Semen collection, characterisation and artificial insemination in the beluga (Delphinapterus leucas) using liquid-stored spermatozoa. Reprod Fertil Dev. 2008;20(7):770-83. http://doi.org/10.1071/RD08031 PMid:18842179.
    » http://doi.org/10.1071/RD08031
  • Ogorevc J, Orehek S, Dovč P. Cellular reprogramming in farm animals: an overview of iPSC generation in the mammalian farm animal species. J Anim Sci Biotechnol. 2016;7(1):10. http://doi.org/10.1186/s40104-016-0070-3 PMid:26900466.
    » http://doi.org/10.1186/s40104-016-0070-3
  • Palomino JM, Mastromonaco GF, Cervantes MP, Mapletoft RJ, Anzar M, Adams GP. Effect of season and superstimulatory treatment on in vivo and in vitro embryo production in wood bison (Bison bison athabascae). Reprod Domest Anim. 2020;55(1):54-63. http://doi.org/10.1111/rda.13580 PMid:31661568.
    » http://doi.org/10.1111/rda.13580
  • Peippo J, Lindeberg H, Mutikainen M, Harmama T-M, Kumpula J, Holand O. In vitro production of the first reindeer (Rangifer tarandus tarandus) blastocysts. In: Proceedings of the 35th annual meeting of the European Embryo Transfer Association; 2019 Sep 12-14; Murcia, Spain. Murcia: AETE; 2019. p. 746.
  • Pennington PM, Marshall KL, Capiro JM, Felton RG, Durrant BS. Ovulation induction in anovulatory southern white rhinoceros (Ceratotherium simum simum) without altrenogest. Conserv Physiol. 2019;7(1):coz033. http://doi.org/10.1093/conphys/coz033 PMid:31249689.
    » http://doi.org/10.1093/conphys/coz033
  • Perry S. Developing assisted reproduction for reptiles, what’s next? Clin Theriogenology. 2021;13:383. https://doi.org/10.58292/ct.v13.9315.
  • Prieto MT, Sanchez-Calabuig MJ, Hildebrandt TB, Santiago-Moreno J, Saragusty J. Sperm cryopreservation in wild animals. Eur J Wildl Res. 2014;60(6):851-64. http://doi.org/10.1007/s10344-014-0858-4
    » http://doi.org/10.1007/s10344-014-0858-4
  • Pukazhenthi BS, Wildt DE. Which reproductive technologies are most relevant to studying, managing and conserving wildlife? Reprod Fertil Dev. 2004;16(1-2):33-46. http://doi.org/10.1071/RD03076 PMid:14972101.
    » http://doi.org/10.1071/RD03076
  • Revive & Restore. [homepage on the internet]. Revive & Restore; 2024. The genetic rescue toolkit; 2024 [cited 2024 March 10]. Available from: https://reviverestore.org/what-we-do/genetic-rescue-toolkit
    » https://reviverestore.org/what-we-do/genetic-rescue-toolkit
  • Robeck TR, Montano GA, Steinman KJ, Smolensky P, Sweeney J, Osborn S, O’Brien JK. Development and evaluation of deep intra-uterine artificial insemination using cryopreserved sexed spermatozoa in bottlenose dolphins (Tursiops truncatus). Anim Reprod Sci. 2013;139(1-4):168-81. http://doi.org/10.1016/j.anireprosci.2013.04.004 PMid:23660366.
    » http://doi.org/10.1016/j.anireprosci.2013.04.004
  • Robeck TR, Steinman KJ, Gearhart S, Reidarson TR, McBain JF, Monfort SL. Reproductive physiology and development of artificial insemination technology in killer whales (Orcinus orca). Biol Reprod. 2004;71(2):650-60. http://doi.org/10.1095/biolreprod.104.027961 PMid:15115725.
    » http://doi.org/10.1095/biolreprod.104.027961
  • Robeck TR, Steinman KJ, Greenwell M, Ramirez K, Van Bonn W, Yoshioka M, Katsumata E, Dalton L, Osborn S, O’Brien JK. Seasonality, estrous cycle characterization, estrus synchronization, semen cryopreservation, and artificial insemination in the Pacific white-sided dolphin (Lagenorhynchus obliquidens). Reproduction. 2009;138(2):391-405. http://doi.org/10.1530/REP-08-0528 PMid:19494046.
    » http://doi.org/10.1530/REP-08-0528
  • Rola LD, Buzanskas ME, Melo LM, Chaves MS, Freitas VJF, Duarte JMB. Assisted reproductive technology in neotropical deer: a model approach to preserving genetic diversity. Animals (Basel). 2021a;11(7):1961. http://doi.org/10.3390/ani11071961 PMid:34209061.
    » http://doi.org/10.3390/ani11071961
  • Rola LD, Zanetti EDS, Del Collado M, Peroni EFC, Duarte JMB. Collection and in vitro maturation of Mazama gouazoubira (brown brocket deer) oocytes obtained after ovarian stimulation. Zygote. 2021b;29(3):216-22. http://doi.org/10.1017/S0967199420000787 PMid:33446301.
    » http://doi.org/10.1017/S0967199420000787
  • Roth TL. That was then, this is now – over two decades of progress in rhinoceros reproductive science and technology. Theriogenology Wild. 2024;4:100065. http://doi.org/10.1016/j.therwi.2023.100065
    » http://doi.org/10.1016/j.therwi.2023.100065
  • Silla AJ, Calatayud NE, Trudeau VL. Amphibian reproductive technologies: approaches and welfare considerations. Conserv Physiol. 2021;9(1):coab011. http://doi.org/10.1093/conphys/coab011 PMid: 33763231.
    » http://doi.org/10.1093/conphys/coab011
  • Sontakke SD. Monitoring and controlling ovarian activities in wild ungulates. Theriogenology. 2018;109:31-41. http://doi.org/10.1016/j.theriogenology.2017.12.008 PMid:29254686.
    » http://doi.org/10.1016/j.theriogenology.2017.12.008
  • Stewart RA, Pelican KM, Crosier AE, Pukazhenthi BS, Wildt DE, Ottinger MA, Howard J. Oral progestin priming increases ovarian sensitivity to gonadotropin stimulation and improves luteal function in the cat. Biol Reprod. 2012;87(6):137. http://doi.org/10.1095/biolreprod.112.104190 PMid:23100619.
    » http://doi.org/10.1095/biolreprod.112.104190
  • Strange A, Alberio R. Review: a barnyard in the lab: prospect of generating animal germ cells for breeding and conservation. Animal. 2023;17(Suppl 1):100753. http://doi.org/10.1016/j.animal.2023.100753 PMid:37567650.
    » http://doi.org/10.1016/j.animal.2023.100753
  • Swanson WF, Penfold LM. Captive wildlife breeding: mammals & birds. In: Skinner MK, editor. Encyclopedia of Reproduction. 2nd ed. Cambridge: Academic Press; 2018. p. 782-90. http://doi.org/10.1016/B978-0-12-809633-8.20631-6
    » http://doi.org/10.1016/B978-0-12-809633-8.20631-6
  • Swegen A, Appeltant R, Williams SA. Cloning in action: can embryo splitting, induced pluripotency and somatic cell nuclear transfer contribute to endangered species conservation? Biol Rev Camb Philos Soc. 2023;98(4):1225-49. http://doi.org/10.1111/brv.12951 PMid:37016502.
    » http://doi.org/10.1111/brv.12951
  • Swengel SR, Tuite ML. Recent advances in scheduling strategies and practical techniques in crane artificial insemination. In: North American Crane Workshop Proceedings; 1997 Jan 10-13; Biloxi, United States. Biloxi: North American Crane; 1997. p. 46-55.
  • Thitaram C, Brown JL. Monitoring and controlling ovarian activity in elephants. Theriogenology. 2018;109:42-7. http://doi.org/10.1016/j.theriogenology.2017.12.009 PMid:29287954.
    » http://doi.org/10.1016/j.theriogenology.2017.12.009
  • Thongphakdee A, Berg DK, Tharasanit T, Thongtip N, Tipkantha W, Punkong C, Tongthainan D, Noimoon S, Maikeaw U, Kajornklin N, Siriaroonrat B, Comizzoli P, Kamolnorranath S. The impact of ovarian stimulation protocol on oocyte quality, subsequent in vitro embryo development, and pregnancy after transfer to recipients in Eld’s deer (Rucervus eldii thamin). Theriogenology. 2017;91:134-44. http://doi.org/10.1016/j.theriogenology.2016.12.021 PMid:28215678.
    » http://doi.org/10.1016/j.theriogenology.2016.12.021
  • Thongphakdee A, Sukparangsi W, Comizzoli P, Chatdarong K. Reproductive biology and biotechnologies in wild felids. Theriogenology. 2020;150:360-73. http://doi.org/10.1016/j.theriogenology.2020.02.004 PMid:32102745.
    » http://doi.org/10.1016/j.theriogenology.2020.02.004
  • Thongphakdee A, Tipkantha W, Punkong C, Chatdarong K. Monitoring and controlling ovarian activity in wild felids. Theriogenology. 2018;109:14-21. http://doi.org/10.1016/j.theriogenology.2017.12.010 PMid:29258698.
    » http://doi.org/10.1016/j.theriogenology.2017.12.010
  • Toosi BM, Tribulo A, Lessard C, Mastromonaco GF, McCorkell RB, Adams GP. Superovulation and embryo transfer in wood bison (Bison bison athabascae). Theriogenology. 2013;80(5):542-51. http://doi.org/10.1016/j.theriogenology.2013.05.020 PMid:23831114.
    » http://doi.org/10.1016/j.theriogenology.2013.05.020
  • United Parks & Resorts. [homepage on the Internet]. United Parks & Resorts; 2024. Species preservation laboratory; 2024 [cited 2024 March 9]. Available from: https://seaworld.org/conservation/species-preservation-laboratory/primary-research/
    » https://seaworld.org/conservation/species-preservation-laboratory/primary-research/
  • Van Eenennaam JP, Linares-Casenave J, Muguet J-B, Doroshov SI. Induced spawning, artificial fertilization, and egg incubation techniques for green sturgeon. N Am J Aquaculture. 2008;70(4):434-45. http://doi.org/10.1577/A07-073.1
    » http://doi.org/10.1577/A07-073.1
  • Wildt DE, Schiewe MC, Schmidt PM, Goodrowe KL, Howard JG, Phillips LG, O’Brien SJ, Bush M. Developing animal model systems for embryo technologies in rare and endangered wildlife. Theriogenology. 1986;25(1):33-51. http://doi.org/10.1016/0093-691X(86)90182-2
    » http://doi.org/10.1016/0093-691X(86)90182-2
  • Wyse J, Latif S, Gurusinghe S, McCormick J, Weston LA, Stephen CP. Phytoestrogens: a review of their impacts on reproductive physiology and other effects upon grazing livestock. Animals (Basel). 2022;12(19):2709. http://doi.org/10.3390/ani12192709 PMid:36230450.
    » http://doi.org/10.3390/ani12192709
  • Zhu H, Shao X, Chen Z, Wei C, Lei M, Ying S, Yu J, Shi Z. Induction of out-of-season egg laying by artificial photoperiod in Yangzhou geese and the associated endocrine and molecular regulation mechanisms. Anim Reprod Sci. 2017;180:127-36. http://doi.org/10.1016/j.anireprosci.2017.03.009 PMid:28342689.
    » http://doi.org/10.1016/j.anireprosci.2017.03.009
  • Zwiefelhofer ML, Shury T, Zwiefelhofer EM, Singh J, Mastromonaco GF, Adams GP. Strategies for oocyte collection and in vitro embryo production in free-roaming bison herds. Conserv Physiol. 2022b;10(1):coac058. http://doi.org/10.1093/conphys/coac058 PMid: 35966755.
    » http://doi.org/10.1093/conphys/coac058
  • Zwiefelhofer ML, Singh J, Zwiefelhofer EM, Mastromonaco GF, Adams GP. Influence of ovarian follicular wave synchronization and single-dose eCG superstimulation on oocyte collection and in vitro embryo production in bison during the ovulatory and anovulatory seasons. Theriogenology. 2022a;187:238-46. http://doi.org/10.1016/j.theriogenology.2022.04.023 PMid:35660373.
    » http://doi.org/10.1016/j.theriogenology.2022.04.023
  • 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
    18 Apr 2024
  • Accepted
    25 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