REVIEW

Transplantation of spermatogonial stem cells in stallions

Heejun Jung1https://orcid.org/0000-0001-9608-1412, Minjung Yoon1,2,*https://orcid.org/0000-0001-9112-1796
Author Information & Copyright
1Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Korea
2Department of Horse, Companion and Wild Animal Science, Kyungpook National University, Sangju 37224, Korea
*Corresponding author: Minjung Yoon, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Korea. Tel: +82-54-530-1233, E-mail: mjyoonemail@gmail.com

© Copyright 2024 Korean Society of Animal Science and Technology. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Aug 30, 2023; Revised: Feb 07, 2024; Accepted: Feb 29, 2024

Published Online: Jul 31, 2024

Abstract

Spermatogonial stem cells originate from gonocytes and undergo self-renewal and differentiation to generate mature spermatozoa via spermatogenesis in the seminiferous tubules of the testis in male mammals. Owing to the unique capacity of these cells, the spermatogonial stem cell transplantation technique, which enables the restoration of male fertility by transfer of germlines between donor and recipient males, has been developed. Thus, spermatogonial stem cell transplantation can be used as an important next-generation reproductive and breeding tool in livestock production. However, in large animals, this approach is associated with many technical limitations and inefficiency. Furthermore, research regrading spermatogonial stem cell transplantation in stallions is limited. Therefore, this review article describes the history and current knowledge regarding spermatogonial stem cell transplantation in animals and challenges in establishing an experimental protocol for successful spermatogonial stem cell transplantation in stallions, which have been presented under the following heads: spermatogonial stem cell isolation, recipient preparation, and spermatogonial stem cell transplantation. Additionally, we suggest that further investigation based on previous unequivocal evidence regarding donor-derived spermatogenesis in large animals must be conducted. A detailed and better understanding of the physical and physiological aspects is required to discuss the current status of this technique field and develop future directions for the establishment of spermatogonial stem cell transplantation in stallions.

Keywords: Spermatogonial stem cells; Transplantation; Germ cells; Rete testis; Stallions

INTRODUCTION

Spermatogenesis is a highly complex process that progressively produces and maintains functional sperm in the seminiferous tubules of the testis throughout the life of male mammals [1]. Spermatogenesis is initiated by the proliferation and differentiation of spermatogonial stem cells (SSCs). They are located adjacent to the basement membrane of the seminiferous tubules of the testis and undergo self-renewal and differentiation to maintain stable populations of spermatogenic cells for the production of mature spermatozoa [2,3]. This unique capacity of SSCs has led to the development of transplantation techniques for use in animal reproduction and regenerative medicine. In contrast to the normal process of spermatogenesis, transplanted donor SSCs migrate from the lumen to the basement membrane of the seminiferous tubules. Before this process, endogenous donor SSCs are isolated from the testicular tissue and transplanted into the recipient’s testis; they pass through the vas deferens, rete testis, and seminiferous tubules, and donor-derived spermatogenesis is re-established in the specific microenvironment, namely niche [4,5].

In 1994, the transplantation technique was first used to produce donor-derived offspring from infertile recipient mice [6]. This report demonstrated that donor-derived spermatogenesis could produce offspring through natural mating and opened up new possibilities for the use of SSCs in biomedicine and agriculture, such as for the production of genetically modified animals, conservation of superior genetic resources, and treatment of male infertility due to injury and disease. The transplantation technique has been extensively studied in various animals, including pigs [79], cattle [1012], goats [1315], shrews [16], monkeys [1719], dogs [20,21], and camels [22]. However, because of species-specific differences, such as anatomical differences, resistance of the lamina propria, and high volume-to-surface ratio, this technique has not been successful in all animals, and only a limited number of studies have demonstrated the production of donor-derived embryos and offspring [8]. A previous study showed that the transplantation technique could not be used in stallions because the rete testis could not be visualized via sonography [23]. To address these issues, we recently conducted a study in which donor germ cells were transplanted directly into the testicular tissue of the recipient’s testes to induce donor-derived spermatogenesis in stallions [24]. However, our modified technique, which involved infusion of donor germ cells into the testicular tissue without using ultrasound guidance, was ineffective in stallions. This finding highlights the importance of using an ultrasound-guided technique and identifying optimal sites for germ cell transplantation in stallions to ensure successful outcomes.

The findings from the stallion study indicated that although the SSC transplantation (SSCT) is promising potential for male fertility restoration in several species, the validation of experimental injection techniques and its effectiveness remains unclear and challenging in the satllions. Thus, detailed procedures and technical improvements need to be developed to achieve higher success rates of SSCT in the stallions.

In this review, we provide a comprehensive overview of the current advancement in the field of SSCT research using several species including large animal models. In particular, it aims to discuss and summarize the SSC biology, ongoing attempts, main limitations, and highlight the requiremnets for the future experimental advancements to improve the SSCT efficiency in the stallions.

Spermatogenesis in stallions

Spermatogenesis is a stem cell-based process in which functional mature spermatozoa are progressively produced by division and differentiation within the seminiferous tubules that compose the testicular tissue. This process is accomplished through three distinct developmental stages, namely spermatocytogenesis, meiosis, and spermiogenesis [25,26]. In stallions, spermatogenesis is typically completed in 57.4 days [27]. The first stage, i.e., spermatocytogenesis, which takes 19.4 days, is initiated by the development of spermatogonia. SSCs are the undifferentiated, immature germ cells adjacent to the basement membrane of the seminiferous tubules and are capable of self-renewal and differentiation for continuous production of mature sperm throughout the adult life [25]. A previous study showed that A1, A2, A3, A4, and B are the differentiating spermatogonial populations in mice and pigs [28], while in cattle and sheep, A1, A2, A3, B1, and B2 were reported to be the differentiating spermatogonial populations [28]. Recently, eight different subtypes of spermatogonia, namely As, Apr, Aal, A1, A2, A3, B1, and B2, have been reported, and As, Apr, Aal spermatogonia are considered SSCs [29]. The two major functions of spermatogonia are duplication by mitosis (A0) to maintain a progenitor population for the spermatogenic lineage and production of daughter cells by mitosis to generate primary spermatocytes that differentiate into spermatozoa [30]. In stallions, the A1 spermatogonia are destined to produce primary spermatocytes. Once they are committed to this developmental line, they divide by mitosis in five stages to form 1 (A1), 2 (A1.2), 4 (A1.4), 8 (A1.8), and eventually 16 potential A2 spermatogonia. The spermatogonia are connected by intercellular bridges to maintain their developmental lineage. Subsequently, the A2 spermatogonia divide by mitosis, first into two A3 spermatogonia, then into two B1 spermatogonia, and finally into two B2 differentiated spermatogonia [31]. Eventually, the B2 spermatogonia divide into two primary spermatocytes that undergo the first meiotic division, which constititutes the second stage of spermatogenesis. In this stage, which also takes 19.4 days for completion, the secondary spermatocyte is formed by division of the primary spermatocyte into two diploid secondary spermatocytes by mitotic division and exchange of genetic material. Two haploid spermatids are then produced from each secondary spermatocyte; this process takes 0.7 days. Spermiogenesis is the final stage of spermatogenesis and is completed in 18.6 days [32]. During this process, spermatids undergo dramatic changes; their nuclei transform, they lose their cytoplasm, and develop tails, thus getting ready to differentiate into mature spermatozoa [33].

Preparation of donor spermatogonial stem cells for transplantation in stallions

The technique of germ cell isolation from testicular tissue is widely used to collect pure individual germ cells. The two-enzyme digestion method is usually applied for isolation in several species, including mouse [34], goat [35], cow [36], buffalo [37], rhesus macaque [38], and pig [39]. This helped in the establishment of a germ cell culture system and determination of various factors affecting spermatogenesis [40]. In addition to germ cell evaluation, it is used to prepare donor germ cells for transplantation into the testis of recipient animals, such as goat [13], boar [41], dog [20], and sheep [42]. The collection of pure germ cells, including SSCs, from the testicular tissue using the two-enzyme digestion technique is essential for successful transplantation because stem cells in donor germ cells are the only cell type that can completely regenerate spermatogenesis in the seminiferous tubules of the recipient testis after transplantation [43]. However, like other adult stem cells, SSCs are present in very small numbers in the testes, and as the testes mature, the number of SSCs decreases. For example, there are only approximately 35,000 SSCs per testis in adult mice, and the percentage of the total number of germ cells in the testis is 0.03% [44]. In neonatal pigs, approximately 7% of the gonocytes were present only in the seminiferous tubules [45]. Thus obtaining large numbers of progenitor cells and other undifferentiated spermatogonia using the isolation technique is difficult [46]. Recently, we evaluated the efficiency of the two-enzyme digestion technique for isolation of the testicular tissue in stallions and demonstrated its effectiveness in isolating high-purity germ cells from the testes [47]. Additionally, specific molecular markers for SSCs, including other germ cells, in the testes of stallions have been developed. Antibodies, such as UTF-1 [48], DAZL [49], PGP9.5 [50], C-kit [51], ACRBP [52], VASA [53], and Lin28 [54], can be used to identify specific germ cells at each developmental stage of spermatogenesis in vitro. In particular, UTF-1 is a specific putative marker for undifferentiated SSCs in stallions [48], and we found that 6.43% of germs cells per 10 g of testicular tissue in prepubertal stallions were positive for UTF-1 [47]. The immature testes are typically used to isolate SSCs for transplantation because they contain a greater number of gonocytes or undifferentiated SSCs than do the mature testes. Nevertheless, this method does not significantly increase the number of isolated SSCs. Hence developing other strategies to enrich the concentration of SSCs to improve the efficiency of germ cell transplantation in stallions is necessary. As an alternative to the use of immature testes, long-term in vitro culture of SSCs is widely known to elevate the number of stem cells [55,56]. Several key growth factors, such as GDNF, GFRa1, bFGF, CSF1, LIF, EGF, and IGF1, which contribute to self-renewal and proliferation of SSCs in vitro have been identified [57,58]. However, the nature of the in vitro culture environment and efficacy of these growth factors on the expansion of SSC numbers in stallions remains to be elucidated. Thus, the next challenge would be to establish an in vitro culture environment to proliferate or differentiate SSCs for transplantation into stallions.

Preparation of recipient for transplantation of donor germ cells in stallions

Preparation of the ideal recipient is a key issue in improving the success rate of SSCT. The fundamental purpose of this step is to suppress all the endogenous germ cells while maintaining the full structure and function of the somatic cells in the seminiferous tubules of the recipient’s testes [59]. This provides the space for development of stem cell niches, in which donor-derived spermatogenesis can be re-established in the basement membrane of the seminiferous tubules in the recipient’s testes after transplantation. Thus, several techniques to deplete endogenous germ cells, such as local glycerol injections [60,61], local irradiation [62,63], heat treatment [64,65], systemic busulfan injections [6,14,66], and the use of genetically defective animals with congenital deletion of endogenous spermatogenesis, have been developed [65]. However, no studies have demonstrated the efficacy of these techniques in depleting endogenous germ cells in stallions. In our previous study, we found that intra-testicular injection of 70% glycerol suppressed some germ cells in the seminiferous tubules in stallions. However, it did not completely deplete endogenous germ cells for transplantation [67]. As an alternative, the chemotherapeutic agent, namely busulfan is widely used to deplete endogenous germ cells to prepare recipient mouse models for transplantation. However, the efficacy of this treatment varies in large animals; moreover, knowledge regarding it is limited owing to the differences in physiological structure, composition, and size between species [18]. Additionally, busulfan is systemically toxic; it causes bone marrow suppression and inhibits hematopoiesis, which occasionally leads to lethal effects. Therefore, the species-specific sublethal dose of busulfan required to deplete endogenous germ cells should be determined. A recent study showed that multiple injections of low concentrations of busulfan can effectively deplete endogenous germ cells without any side effects in stallions [66]. However, some problems with transplantation persist. Although treatment with busulfan is more effective than that with glycerol, it cannot completely deplete germ cells, and recovery of the endogenous germ cells is inevitable. This eventually disrupts the re-establishment of transplanted donor SSCs. Therefore, along with depletion of endogenous germ cells of the recipient, determining the optimal time period for regeneration of donor-derived spermatogenesis is important for successful transplantation in stallions.

Donor germ cell transplantation into recipient stallion testes

Three methods are typically employed to ensure sufficient engraftment of donor SSCs into the lumen of seminiferous tubules of the recipient’s testes. In mice, donor SSCs have been successfully transferred into the seminiferous tubules of the recipient’s testes via efferent duct injection [6]. The efferent ducts are directly connected to the rete tubules and epididymis of the testes. The seminiferous tubules converge to form the rete tubules, which carry mature sperm from the seminiferous tubules to the efferent ducts [68]. Thus, efferent duct injection is effective in transferring donor SSCs into the seminiferous tubules of the recipient’s testes. However, this transplantation technique, which is commonly used in rodents, cannot be applied to all animals because of differences in anatomical structures, greater resistance of the lamina propria, and large volume-to-surface ratio [18]. In mammals, the highly coiled, tightly packed seminiferous tubules are lined with individual compartments consisting of inwardly extending tunica albuginea [46]. Therefore, the potential rise in intratubular pressure during transplantation may rupture the seminiferous tubules and membrane of the testes [68]. In bovine animals, donor SSCs are directly injected into the seminiferous tubules of the recipient’s testes; however, this method was found to be difficult and inefficient because of the highly convoluted tubular structure of the bovine testes. The rete testis has a distinct structure and color and can be easily distinguished from the surrounding tissues in the testicles, thus allowing more practical access to the seminiferous tubules [7]. Hence a large volume of germ cell suspension can be injected into the rete testis, thereby improving the efficiency of cell transfer and success rate of transplantation. However, the structural location of the rete testis varies between species. Unlike the rete testis of rodents, such as mice, rats, and hamsters, which is located closer to the subcapsular area of the testis, that of most other species, such as cats, dogs, bulls, boars, and monkeys, is located deep in the center of the testis [4, 68, 69]. Hence, transplantation of SSCs through the rete testis is more difficult and less feasible in large animals than it is in rodent models. To overcome this problem, a method to inject donor SSCs into the rete testis of large animals under ultrasound guidance was developed. However, a previous study showed that the rete testis cannot be identified in stallions using sonography [23]. Therefore, instead of injection into the rete testis, 5 mL of iodixanol-contrasted fluorescein solution was injected into the parenchyma below the epididymis, which helped identify contrast in the vessels extending toward the head of the epididymis. Thus, this previous finding indicated that the testicular injection method may be applicable for SSCT in stallions. Hence, we transplanted SSCs directly into the testicular parenchyma of stallions without identifying the rete testis by sonography; however, we found no donor-derived sperm in the recipient semen after transplantation. This approache revealed an additional issue that the recipient pose and anesthesia should be considered for the successful transplantation. Because since the recipient horse was standing without anesthesia while in performing transplantation, even if the needles were properly inserted into the testicular parenchyma, the needle position and germ cell migration might be interrupted by continuous movement of the recipient. However, the general anesthesia in horses carries a mortality rate 1 in 10 due to they have heavy muscles and organs, which do not function while standing, as well as low blood oxygen levels, which can also cause problems. Moreover, the expertise in anesthesia, appropriate drugs, and sepcific equipments, along with a suitable environment, are required. Thus, the testicular injection of donor SSCs without considering the pose and anesthesia of the recipient is not an optimal transplantation techniques for producing donor-derived sperm [24]. Therefore, alternative approach must be considered to obtain donor sperm without transplanting germ cells into the testes of recipient stallions. Several techniques, such as in vitro single germ cell culture [70], testicular tissue culture [71,72], xenograft [73], and xenotransplantation [74,75], have been introduced to produce donor sperm without transplanting SSC in recipient testis. However, their efficacy in stallions has not yet been confirmed. Overall, the current experimental approaches used to produce donor SSC-derived sperm in stallions are inefficient, and clarity regarding the alternative methods is lacking. Additionally, factors such as enrichment of donor SSCs and preparation of germ cells with mitotically active Sertoli cells must be considered to ensure successful transplantation. Finally, alternative approaches to obtain donor sperm without transplanting germ cells into the testes of recipient stallions are required.

CONCLUSION

According to recent research in the field of animal breeding, alternative and innovative breeding techniques are being widely explored to efficiently breed animals with superior genetics. These techniques include artificial insemination (AI), somatic cell nuclear transfer technology (SCNT), and SSCT. A concept that is being actively investigated in this field of research is the utilization of feeder mermaphrodite-based SSC. The aim of this approach is to breed and disseminate superior biological genes in males and females without relying on female animals. This greatly accelerates livestock breeding and production. However, several key factors, including long-term in vitro culture and large-scale proliferation of SSCs, development of ideal recipient models, and effective cell migration and migration pathways, must be addressed. While SSCT techniques for use in large animals are currently available, obtaining successful results remains challenging. Therefore, further studies are required to establish a reliable, efficient, and productive SSCT protocol for large animals. NANOS2 gene knock-out studies have primarily been performed using experimental animals, such as mice, but recently, successful cases have been reported in larger animals. The infertile males generated through this gene-editing technology have the capacity to successfully undergo SSCT. Hence, developing technologies to discover and manipulate various genes involved in male germline maintenance to develop the ideal recipient animals is necessary. Furthermore, research regarding genetic modification of SSCs should be accelerated to overcome high costs and improve the efficiency of pronuclear injection and embryo-based genome modification techniques, including SCNT. We suggested that this review may provide guidelines for future studies of SSC transplantation in the large animals that have not yet been performed.

Competing interests

No potential conflict of interest relevant to this article was reported.

Funding sources

Not applicable.

Acknowledgements

Not applicable.

Availability of data and material

Upon reasonable request, the datasets of this study can be available from the corresponding author.

Authors’ contributions

Conceptualization: Yoon M.

Writing - original draft: Jung H.

Writing - review & editing: Jung H, Yoon M.

Ethics approval and consent to participate

This article does not require IRB/IACUC approval because there are no human and animal participants.

REFERENCES

1.

Griswold MD. Spermatogenesis: the commitment to meiosis. Physiol Rev. 2016; 96:1-17

2.

Smith LB, Walker WH. The regulation of spermatogenesis by androgens. Semin Cell Dev Biol. 2014; 30:2-13

3.

Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod. 2018; 99:52-74

4.

Ogawa T, Aréchaga JM, Avarbock MR, Brinster RL. Transplantation of testis germinal cells into mouse seminiferous tubules. Int J Dev Biol. 1997; 41:111-22

5.

Nagano M, Avarbock MR, Brinster RL. Pattern and kinetics of mouse donor spermatogonial stem cell colonization in recipient testes. Biol Reprod. 1999; 60:1429-36

6.

Brinster RL, Zimmermann JW. Spermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci USA. 1994; 91:11298-302

7.

Honaramooz A, Megee SO, Dobrinski I. Germ cell transplantation in pigs. Biol Reprod. 2002; 66:21-8

8.

Zeng W, Tang L, Bondareva A, Honaramooz A, Tanco V, Dores C, et al. Viral transduction of male germline stem cells results in transgene transmission after germ cell transplantation in pigs. Biol Reprod. 2013; 88:27

9.

Kim BG, Kim YH, Lee YA, Kim BJ, Kim KJ, Jung SE, et al. Production of transgenic spermatozoa by lentiviral transduction and transplantation of porcine spermatogonial stem cells. Tissue Eng Regen Med. 2014; 11:458-66

10.

Izadyar F, Den Ouden K, Stout TA, Stout J, Coret J, Lankveld DP, et al. Autologous and homologous transplantation of bovine spermatogonial stem cells. Reproduction. 2003; 126:765-74

11.

Herrid M, Vignarajan S, Davey R, Dobrinski I, Hill JR. Successful transplantation of bovine testicular cells to heterologous recipients. Reproduction. 2006; 132:617-24

12.

Stockwell S, Herrid M, Davey R, Brownlee A, Hutton K, Hill JR. Microsatellite detection of donor-derived sperm DNA following germ cell transplantation in cattle. Reprod Fertil Dev. 2009; 21:462-8

13.

Honaramooz A, Behboodi E, Megee SO, Overton SA, Galantino-Homer H, Echelard Y, et al. Fertility and germline transmission of donor haplotype following germ cell transplantation in immunocompetent goats. Biol Reprod. 2003; 69:1260-4

14.

Brinster RL, Avarbock MR. Germline transmission of donor haplotype following spermatogonial transplantation. Proc Natl Acad Sci USA. 1994; 91:11303-7

15.

Honaramooz A, Megee S, Zeng W, Destrempes MM, Overton SA, Luo J, et al. Adeno-associated virus (AAV)-mediated transduction of male germ line stem cells results in transgene transmission after germ cell transplantation. FASEB J. 2008; 22:374-82

16.

Li CH, Yan LZ, Ban WZ, Tu Q, Wu Y, Wang L, et al. Long-term propagation of tree shrew spermatogonial stem cells in culture and successful generation of transgenic offspring. Cell Res. 2017; 27:241-52

17.

Schlatt S, Foppiani L, Rolf C, Weinbauer GF, Nieschlag E. Germ cell transplantation into X-irradiated monkey testes. Hum Reprod. 2002; 17:55-62

18.

Schlatt S, Rosiepen G, Weinbauer GF, Rolf C, Brook PF, Nieschlag E. Germ cell transfer into rat, bovine, monkey and human testes. Hum Reprod. 1999; 14:144-50

19.

Jahnukainen K, Ehmcke J, Quader MA, Saiful Huq M, Epperly MW, Hergenrother S, et al. Testicular recovery after irradiation differs in prepubertal and pubertal non-human primates, and can be enhanced by autologous germ cell transplantation. Hum Reprod. 2011; 26:1945-54

20.

Kim Y, Turner D, Nelson J, Dobrinski I, McEntee M, Travis AJ. Production of donor-derived sperm after spermatogonial stem cell transplantation in the dog. Reproduction. 2008; 136:823-31

21.

Harkey MA, Asano A, Zoulas ME, Torok-Storb B, Nagashima J, Travis A. Isolation, genetic manipulation, and transplantation of canine spermatogonial stem cells: progress toward transgenesis through the male germ-line. Reproduction. 2013; 146:75-90

22.

Herrid M, Nagy P, Juhasz J, Morrell JM, Billah M, Khazanehdari K, et al. Donor sperm production in heterologous recipients by testis germ cell transplantation in the dromedary camel. Reprod Fertil Dev. 2019; 31:538-46

23.

Smith E. Spermatogonial stem cell transfer to a mule [Master’s thesis]. Murfreesboro, TN: Middle Tennessee State University. 2015

24.

Jung H, Yoon M. Germ cell transplantation in stallion testes. J Equine Vet Sci. 2021; 106:103748

25.

Oatley JM, Brinster RL. The germline stem cell niche unit in mammalian testes. Physiol Rev. 2012; 92:577-95

26.

de Rooij DG. The nature and dynamics of spermatogonial stem cells. Development. 2017; 144:3022-30

27.

Swierstra EE, Gebauer MR, Pickett BW. Reproductive physiology of the stallion: I. spermatogenesis and testis composition. J Reprod Fertil. 1974; 40:113-23

28.

França LR, Avelar GF, Almeida FFL. Spermatogenesis and sperm transit through the epididymis in mammals with emphasis on pigs. Theriogenology. 2005; 63:300-18

29.

Costa GMJ, Avelar GF, Rezende-Neto JV, Campos-Junior PHA, Lacerda SMSN, Andrade BSC, et al. Spermatogonial stem cell markers and niche in equids. PLOS ONE. 2012; 7e44091

30.

Johnson L, Blanchard TL, Varner DD, Scrutchfield WL. Factors affecting spermatogenesis in the stallion. Theriogenology. 1997; 48:1199-216

31.

Johnson L. Seasonal differences in equine spermatocytogenesis. Biol Reprod. 1991; 44:284-91

32.

Morel MCGD. Equine reproductive physiology, breeding and stud management. 4th ed Wallingford, Oxfordshire: CABI. 2015

33.

Holstein AF, Schulze W, Davidoff M. Understanding spermatogenesis is a prerequisite for treatment. Reprod Biol Endocrinol. 2003; 1:107

34.

Chang YF, Lee-Chang JS, Panneerdoss S, MacLean JA, Rao MK. Isolation of Sertoli, Leydig, and spermatogenic cells from the mouse testis. BioTechniques. 2011; 51:341-4

35.

Heidari B, Rahmati-Ahmadabadi M, Akhondi MM, Zarnani AH, Jeddi-Tehrani M, Shirazi A, et al. Isolation, identification, and culture of goat spermatogonial stem cells using c-kit and PGP9.5 markers. J Assist Reprod Genet. 2012; 29:1029-38

36.

Izadyar F, den Ouden K, Creemers LB, Posthuma G, Parvinen M, de Rooij DG. Proliferation and differentiation of bovine type A spermatogonia during long-term culture. Biol Reprod. 2003; 68:272-81

37.

Rafeeqi T, Kaul G. Isolation and enrichment of type A spermatogonia from pre-pubertal buffalo (Bubalus bubalis) testis. Andrologia. 2013; 45:195-203

38.

Hermann BP, Sukhwani M, Lin CC, Sheng Y, Tomko J, Rodriguez M, et al. Characterization, cryopreservation, and ablation of spermatogonial stem cells in adult rhesus macaques. Stem Cells. 2007; 25:2330-8

39.

Lee KH, Lee WY, Kim JH, Yoon MJ, Kim NH, Kim JH, et al. Characterization of GFRα-1-positive and GFRα-1-negative spermatogonia in neonatal pig testis. Reprod Domest Anim. 2013; 48:954-60

40.

Dores C, Alpaugh W, Dobrinski I. From in vitro culture to in vivo models to study testis development and spermatogenesis. Cell Tissue Res. 2012; 349:691-702

41.

Mikkola M, Sironen A, Kopp C, Taponen J, Sukura A, Vilkki J, et al. Transplantation of normal boar testicular cells resulted in complete focal spermatogenesis in a boar affected by the immotile short-tail sperm defect. Reprod Domest Anim. 2006; 41:124-8

42.

Herrid M, Olejnik J, Jackson M, Suchowerska N, Stockwell S, Davey R, et al. Irradiation enhances the efficiency of testicular germ cell transplantation in sheep. Biol Reprod. 2009; 81:898-905

43.

Kanatsu-Shinohara M, Morimoto H, Shinohara T. Enrichment of mouse spermatogonial stem cells by melanoma cell adhesion molecule expression. Biol Reprod. 2012; 87:139

44.

Tagelenbosch RAJ, de Rooij DG. A quantitative study of spermatogonial multiplication and stem cell renewal in the C3H/101 F1 hybrid mouse. Mutat Res. 1993; 290:193-200

45.

Honaramooz A, Behboodi E, Hausler CL, Blash S, Ayres S, Azuma C, et al. Depletion of endogenous germ cells in male pigs and goats in preparation for germ cell transplantation. J Androl. 2005; 26:698-705

46.

Zhao X, Wan W, Zhang X, Wu Z, Yang H. Spermatogonial stem cell transplantation in large animals. Animals. 2021; 11:918

47.

Jung H, Yoon M. Isolation of germ cells from testes of stallions using collagenase and trypsin-ethylenediaminetetraacetic acid. J Equine Vet Sci. 2016; 43:82-7

48.

Jung H, Roser JF, Yoon M. UTF1, a putative marker for spermatogonial stem cells in stallions. PLOS ONE. 2014; 9e108825

49.

Jung HJ, Song H, Yoon MJ. Stage-dependent DAZL localization in stallion germ cells. Anim Reprod Sci. 2014; 147:32-8

50.

Choi Y, Jung Y, Kim S, Kim J, Jung H, Yoon M. Stage-dependent expression of protein gene product 9.5 in donkey testes. Animals. 2020; 10:2169

51.

Jung H, Song H, Yoon M. The KIT is a putative marker for differentiating spermatogonia in stallions. Anim Reprod Sci. 2015; 152:39-46

52.

Kim JT, Jung HJ, Song H, Yoon MJ. Acrosin-binding protein (ACRBP) in the testes of stallions. Anim Reprod Sci. 2015; 163:179-86

53.

Kim JY, Jung HJ, Yoon MJ. VASA (DDX4) is a putative marker for spermatogonia, spermatocytes and round spermatids in stallions. Reprod Domest Anim. 2015; 50:1032-8

54.

Lee G, Jung H, Yoon M. The Lin28 expression in stallion testes. PLOS ONE. 2016; 11e0165011

55.

Kanatsu-Shinohara M, Miki H, Inoue K, Ogonuki N, Toyokuni S, Ogura A, et al. Long-term culture of mouse male germline stem cells under serum-or feeder-free conditions. Biol Reprod. 2005; 72:985-91

56.

Kanatsu-Shinohara M, Ogonuki N, Inoue K, Miki H, Ogura A, Toyokuni S, et al. Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod. 2003; 69:612-6

57.

Zhang P, Chen X, Zheng Y, Zhu J, Qin Y, Lv Y, et al. Long-term propagation of porcine undifferentiated spermatogonia. Stem Cells Dev. 2017; 26:1121-31

58.

Zheng Y, Tian X, Zhang Y, Qin J, An J, Zeng W. In vitro propagation of male germline stem cells from piglets. J Assist Reprod Genet. 2013; 30:945-52

59.

Brinster CJ, Ryu BY, Avarbock MR, Karagenc L, Brinster RL, Orwig KE. Restoration of fertility by germ cell transplantation requires effective recipient preparation. Biol Reprod. 2003; 69:412-20

60.

Igdoura SA, Wiebe JP. Suppression of spermatogenesis by low-level glycerol treatment. J Androl. 1994; 15:234-43

61.

Wiebe JP, Barr KJ. The control of male fertility by 1,2,3-trihydroxypropane (THP; glycerol): rapid arrest of spermatogenesis without altering libido, accessory organs, gonadal steroidogenesis, and serum testosterone, LH and FSH. Contraception. 1984; 29:291-302

62.

Creemers LB, Meng X, den Ouden K, van Pelt AMM, Izadyar F, Santoro M, et al. Transplantation of germ cells from glial cell line-derived neurotrophic factor-overexpressing mice to host testes depleted of endogenous spermatogenesis by fractionated irradiation. Biol Reprod. 2002; 66:1579-84

63.

Shuttlesworth GA, De Rooij DG, Huhtaniemi I, Reissmann T, Russell LD, Shetty G, et al. Enhancement of A spermatogonial proliferation and differentiation in irradiated rats by gonadotropin-releasing hormone antagonist administration. Endocrinology. 2000; 141:37-49

64.

Ma W, Wang J, Gao W, Jia H. The safe recipient of SSC transplantation prepared by heat shock with busulfan treatment in mice. Cell Transplant. 2018; 27:1451-8

65.

Ma W, An L, Wu Z, Wang X, Guo M, Miao K, et al. Efficient and safe recipient preparation for transplantation of mouse spermatogonial stem cells: pretreating testes with heat shock. Biol Reprod. 2011; 85:670-7

66.

Jung H, Yoon M. Effects of intravenous multiple busulfan injection on suppression of endogenous spermatogenesis in recipient stallion testes. J Anim Sci Technol. 2021; 63:1194-203

67.

Jung H, Yoon M. Effects of intratesticular injection of 70% glycerin of stallions. J Equine Vet Sci. 2017; 49:1-10

68.

Gul M, Hildorf S, Dong L, Thorup J, Hoffmann ER, Jensen CFS, et al. Review of injection techniques for spermatogonial stem cell transplantation. Hum Reprod Update. 2020; 26:368-91

69.

Dym M. The mammalian rete testis — a morphological examination. Anat Rec. 1976; 186:493-523

70.

Kim YH, Kang HG, Kim BJ, Jung SE, Karmakar PC, Kim SM, et al. Enrichment and in vitro culture of spermatogonial stem cells from pre-pubertal monkey testes. Tissue Eng Regen Med. 2017; 14:557-66

71.

Baert Y, Braye A, Struijk RB, van Pelt AMM, Goossens E. Cryopreservation of testicular tissue before long-term testicular cell culture does not alter in vitro cell dynamics. Fertil Steril. 2015; 104:1244-52.E4

72.

Reda A, Hou M, Winton TR, Chapin RE, Söder O, Stukenborg JB. In vitro differentiation of rat spermatogonia into round spermatids in tissue culture. Mol Hum Reprod. 2016; 22:601-12

73.

Honaramooz A, Snedaker A, Boiani M, Schöler H, Dobrinski I, Schlatt S. Sperm from neonatal mammalian testes grafted in mice. Nature. 2002; 418:778-81

74.

Ogawa T, Dobrinski I, Avarbock MR, Brinster RL. Xenogeneic spermatogenesis following transplantation of hamster germ cells to mouse testes. Biol Reprod. 1999; 60:515-21

75.

Clouthier DE, Avarbock MR, Maika SD, Hammer RE, Brinster RL. Rat spermatogenesis in mouse testis. Nature. 1996; 381:418-21