Open access peer-reviewed chapter

Sperm Cryopreservation in Farm Animals Using Nanotechnology

Written By

Muhammad Faheem Akhtar and Changfa Wang

Submitted: 13 December 2022 Reviewed: 15 December 2022 Published: 16 May 2023

DOI: 10.5772/intechopen.1001473

From the Edited Volume

Equine Science - Applications and Implications of New Technologies

Juan Carlos Gardón Poggi and Katy Satué Ambrojo

Chapter metrics overview

109 Chapter Downloads

View Full Metrics

Abstract

Sperm cryopreservation is one premier biotechnology in assisted reproduction. In recent decades, there seemed to be an increasing trend in the usage of cryopreserved semen in the equine industry. Post-thaw semen quality and values are not the same, even in different equine species. Similarly, there are species-specific alterations in sperm physiology, i.e., sperm head, kinetic properties, plasma membrane integrity (PMI), and freezability. Albeit, the viability of sperm varies in the female reproductive tract in mares, jennies, and ponies. The absence of standardized methodology in various steps of sperm cryopreservation, i.e., male health examination, semen collection, dilution, semen centrifugation, and pre-and post-thaw semen quality analysis, results in variations in opinions. As compared to other farm animals, assisted reproductive technologies (A.R.T.) are not applied to the same extent in equines. This chapter aims to provide an update on sperm cryopreservation in equine.

Keywords

  • sperm cryopreservation
  • cryodamage
  • equine and other farm animals
  • nanotechnology
  • artificial insemination

1. Introduction

In 2022, the world population will elevate to 8 billion and rise to 9.7 billion by 2050—this massive surge in demands exploring sublime protein sources both from animals and plants [1]. Optimized equine production can be achieved by focusing on all aspects, including epidemiology, nutrition, management, and reproduction. To optimize equine production from farm animals, including horses, donkeys, ponies, etc., the application of biotechnology tools in copy is inevitable. For the better expansion of the equine industry, the semen quality of equine male stock and the causes of low conception rate in female equine stock needs dire attention.

Equine products (meat, milk, skin) are quite popular in European countries and China. Donkey skin and its byproducts are exported from Brazil to China [2]. In the past and even now, the donkey is considered a free-ranging animal, and less attention was given to its nutrition, welfare, and, most importantly, reproduction. Seasonality in reproduction makes equine more challenging for the application of reproductive biotechnologies. But the good news is that, with time and awareness, the current scenario is changing, and marvelous achievements have been made by focusing on assisted reproductive technologies, including artificial insemination (A.I.), sperm cryopreservation, and embryo transfer.

The exploration of cryoprotective agents proved to be a landmark in semen cryopreservation and, ultimately, farm animal reproduction [3, 4, 5]. Cryopreservation is a superb tool for conserving animal genetic resources. Endangered animal breeds can also be preserved by cryopreserving their semen and producing genetically improved animals. Female breeder stock in estrus can be inseminated anytime with cryopreserved semen. Semen quality and cryotolerance vary among different animal species [6]. There are several steps involved in semen cryopreservation, during which semen quality also lowers. Various studies elaborate on maintaining sperm structure and functionality during cryopreservation by enhancing the cryoprotectant’s concentration [7]. Sperm are sensitive to temperature fluctuation during cryopreservation [8]. In this scenario, the quality of post-thaw semen needs to be addressed. Cryopreservation alters acrosome integrity and mitochondrial activity and enhances reactive oxygen species (R.O.S) production.

As a consequence, the integrity of the nucleoprotein structure declines. So, different molecular alterations following sperm cryopreservation are inevitable to explore because one ejaculate is composed of millions of sperms. The fusion of sperm and egg is essential for fertilization [9]. Various breeds of donkeys are kept worldwide, and the trend of producing hybrid donkeys is increasing with time in many countries [10]. For example, a mating horse and donkey hybrid resulted in a highly desirable animal that combined the best features of both animals [11]. In the coming parts of the chapter, we will elaborate, step by step, on different structural and molecular changes in sperm during semen cryopreservation. Research involving donkey semen needs dire attention [12]. The fertility rates of frozen donkey semen are (0–36%) [13]. The optimum conception rate of 61.5% has been achieved till now for jennies artificially inseminated with donkey cryopreserved semen [14]. Reproductive biotechnologies applied in equine, especially donkeys, still need comprehensive research. In the following sections, we will discuss step by step various molecular alterations in semen cryopreservation across multiple farm animals including equine.

Advertisement

2. Sperm cryopreservation cause alterations in the cell membrane

There are several advantages of semen freezing, but certain disadvantages are also there. In equine, abrupt temperature alterations during semen freezing and thawing damage sperm cells resulting in cell membrane changes and even cell mortality up to 10–50% [15]. Cryoprotectants upgrade the ability of sperm to bear temperature fluctuation without affecting their functionality [16]. Thermal stress forms crystals inside sperm cells resulting in cryodamage [17]. In sperm cells, crystals are formed due to imbalanced extracellular and intracellular solute contents [18]. The extracellular medium upgrades sperm volume by passive diffusion [19]. During semen freezing and thawing, these factors affect lipid-protein complexes in Chinese hamsters [20]. They were lowering temperature results in protein adhesion. This process weakens sperm plasmalemma and enhances its permeability, ultimately decreasing sperm metabolism in poultry [21].

Advertisement

3. Molecular alterations after sperm cryopreservation

Molecular alterations after sperm cryopreservation in equine results in lowered spermatozoa longevity. In horses, such a situation is more problematic due to prolonged estrus, and the timing of ovulation requires more careful diagnoses for applying artificial insemination (AI). Regular ultrasound examinations are necessary for mares, and ovulation induction is unavoidable [22]. Significant differences in the reproductive biology of equine, bovine, poultry, and other animals affect sperm cryopreservation; e.g., the average ejaculate volume of poultry ranges from 0.1–0.3 mL as compared to 5–8 mL in the bull. In eutherian mammals, inside sperm chromatin, the type of protamines (P1 and P2) affects cryodamage [23]. Protamines (P1 and P2) alter in various animal species [24]. Inside chromatin, ROS production, and mechanical stress affect the DNA integrity of sperm [25, 26]. Cryopreservation lowers sperm messenger ribonucleic acid (mRNA), ultimately impairing its function [27]. Fertility of bull thawed bull semen is higher with elevated levels of mRNA’s AK1, IB5, TIMP, SNRPN2, and PLCz1 [28]. In oocytes, mRNA aids in the production of proteins during embryogenesis [29]. Sperms cannot replace the lost mRNA during cryopreservation [30]. Epigenetic factors affect freezing and thawing. All these factors collectively are involved in gene expression levels, post-translational histone alterations, chromatin remodeling, DNA methylation, and non-coding RNAs [31, 32]. The frozen, thawed bull semen had a differential abundance of 86 microRNAs [33]. In pig fresh and frozen semen, 135 miRNAs elaoborate metabolic pathways [34]. So, cryopreservation changes apoptotic genes, mitochondrial membrane, DNA fragmentation, phosphatidylserine externalization, and caspase activation [35, 36].

Advertisement

4. Impact of sperm cryopreservation on embryo

Application of assisted reproductive technologies (ART) in the horse is scarce and is only reported in vivo matured oocytes [37]. In vitro, matured oocytes were inseminated in mares, and embryos were generated [37]. Sperm DNA methylation is an inevitable aspect of embryo development, and DNA methylation is directly affected by freezing and thawing procedures, ultimately affecting embryo development [38]. This process involves connecting the methyl group to the cytosines of CpG regions [38]. In horse sperm, DNA methylation improves after cryopreservation (0.6% in fresh and 5.4% in thawed semen) [39]. Abnormal DNA methylation during cryopreservation is why fertilization fails during artificial insemination. Cryopreservation affects early embryonic development. Oocyte inherits epigenetics from nucleosomes and methylated DNA during fertilization [40]. Embryo development is affected by transcription factors [41]. After AI with frozen-thawed semen, downregulation of transcription factors was observed in horse embryos. Transcription factors were downregulated after AI in equine [42]. Hindrance during embryonic development is connected to lowered transcription factors, i.e., TCF7L1, BTEB3, CPBP, KLF3, and NF-1 [41]. In bovine, after cryopreservation, the appraisal of fresh and frozen-thawed semen showed variant mRNA and miRNA profiles [36]. mRNAs’ maternal functions can be controlled by sperm-born miRNAs [43]. The mRNA profiles of embryos can be changed by sperm cryopreservation [44]. Similar variations were noticed in frozen, thawed swine semen after adding glycerol as a cryoprotectant [45].

Advertisement

5. Redox imbalance and its effect on mitochondria

The balance between ROS production and the antioxidant defense mechanism is inevitable in cryopreservation. Otherwise, it leads to oxidative stress. Oxygen free radicals, i.e., hydrogen peroxide (H2O2), superoxide anion (O2−), and hydroxyl radical (OH−), are part of ROS [46]. ROS are critical for sperm function. Sperm cryopreservation triggers apoptotic pathways, while ROS concentrations elevate the optimal value [47]. Increased ROS concentration structurally damages the sperm’s DNA, ultimately disrupting fertility [48]. During cryopreservation, ROS production modifies sperm’s mitochondrial membrane potential [35]. Static oxidative reducing potential (sORP) influences the redox balance in cryopreserved stallion semen [30]. The inclusion of Rosiglitazone can upgrade mitochondrial membrane activity in cryopreserved equine semen. It lowers caspase-3 activity and eventually halts the activation of apoptotic pathways. The addition of Rosiglitazone can aid to acquire redox balance in cryopreservation. In equine semen, rosiglitazone addition aids in AKT (kinase B) protein phosphorylation, which aids in maintaining a balance between apoptotic pathways and cell survival [30]. Compared to other farm animals, ROS production is well reported in equine. ROS production in the mitochondria is scarce in swine before freezing [49]. In swine, the effect of ROS on mitochondrial membrane integrity and lipid peroxidation is better in fresh semen compared to frozen-thawed semen [50]. In equine, lipid peroxidation (LPO) is a primary factor in sperm cryodamage [37]. Mitochondria is a significant source of oxidative stress to spermatozoa [51]. ROS production is elevated in sperm mitochondria during freezing-thawing, while the osmotic mechanism may upgrade mitochondrial membrane permeability, resulting in apoptosis [52].

Advertisement

6. Impact on sperm mobility

Sperm head, size, and movement are disrupted by cryopreservation. In humans, cryopreservation applies oxidative stress on sperm, resulting in cryodamage and reducing progressive motility [53]. Cryopreservation affects the lipids of sperm’s membrane, survival rate, and motility and harms DNA in humans [53]. After cryopreservation, sperm motility is notably affected [53]. Computer-assisted sperm analysis (CASA) aids in evaluating sperm kinetics [54]. Morphological changes in sperm can be analyzed with a phase contrast microscope camera attached to the computer screen. Sperms’ waggling movement can be recorded in sequential images [55]. CASA aids in analyzing different salient factors, e.g., oscillation, straightness, and linearity. It enunciates the frequency the sperm head movement [56]. Sperm can be classified based on their motility by using the CASA system [57]. The categorizing of sperm based on motility and morphology determined various aspects for elaborating sperm biology; sperm motility is influenced by thawing and freezing in bulls, stallions, rams, and boars [55]. In some previous research, sperm motility was connected with fertilizing capacity and viability [57]. Sperm cryopreservation affects sperm motility in equine also, and cryopreserved semen is less frequently used as compared to fresh semen [58]. Fertilizing capacity of frozen-thawed semen varies among various stallion species [59].

In bovine, sperm populations have quick and non-linearity in movements that are elevated in post-thaw sperm activity [60]. Cryopreservation can amend the plasma membrane of sperm [61, 62]. Table 1 enunciates the alterations in sperm cryopreservation in different farm animals and various factors.

Specie.PM (%)AI (%)MF (%)ROS (%)Sperm M & V (%)References
Bovine4010–19154850[63]
Porcine503030260[64]
Ovine8050301.530–40[65]
Equine701235130[66]
Caprine68–7373–81N/AN/AN/A[67]

Table 1.

Sperm cryopreservation variations are specific to species.

Abbreviations: PM (%), progressive motility; A.I (%), acrosome integrity; M.F. (%) mitochondrial function; R.O.S. (%) reactive oxygen species; Sperm M&V (%), sperm motility and viability.

Advertisement

7. Markers of freezability

The composition of cryopreserved semen varies among the same species of sires and between the same ejaculates [18]. Different types of stress, like osmotic and thermal stress, dominate while thawing and freezing procedures and affect sperm response. Seminal plasma (SP) proteins affect sperm’s ability in cattle, pigs, equine, and sheep [68]. Sperm cryopreservation is affected by these SP proteins as they diversify from individual to individual [69]. During cryopreservation, these proteins go through carbonylation, which helps in oxidation; as a conclusion malfunctioning and alterations of protamine of SP proteins occur. These proteins withstand oxidative stress [49, 70]. AKAP4 and proAKAP4 proteins uncover cryodamage in ovine, equine, and porcine [71]. High quantities of HSP90AA1 and HSPA8 proteins enhance sperm cryopreservation chances in bovine [72, 73]. Upgraded post-thaw sperm viability and motility were observed after an elevated concentration of HSP90AA1 protein in swine. Likewise, inside the cell plasma membrane, aquaporins control the permeability of water and cryoprotectants during cryopreservation [74, 75]. In swine and bovine, AQP3 and AQP7 proteins affect sperm cry tolerance. Another crucial protein for sperm cryopreservation in pigs is VDAC2 [76]. Also, an upgraded amount of GSTM3 in swine sperm enunciates higher freezibility after cryopreservation [77]. The improved concentration of SP proteins aids in Cryotolerance. Fibronectin-1 (FN1) helps the thawing and freezing processes in boars [78]. The proteins TCP-1 and 26 S proteasome in swine promote sperm cryopreservation [79].

Advertisement

8. Impact of cryoprotectants

Cell metabolism is downregulated while storing various tissues and cells. The primary aim for sublime sperm functionality is to provide optimum pH, temperature, and osmolarity. In humans, cryoprotective media reduce ice crystal formation and cold shock, resulting in sperm functionality [80]. Permeable and non-permeable cryoprotectants have 4–5% glycerol and 20% egg yolk that guard against cryodamage. Sperm’s lipid bilayer is protected by a plasma membrane combined with egg yolk’s low-density lipoproteins (LDL) [81].

Addition of low-density lipoproteins (LDL) elevated sperm post-thaw quality in cattle [82], pigs [83], sheep [84], and horses [51]. During cryopreservation, a concentration of 9% LDL lowers sperm DNA damage in pigs [83]. Researchers and scientists are looking to substitute egg yolk as a cryoprotectant due to its bacterial contamination [85]. Soy lecithin is a vital substitute LDL having identical functionality [86]. Alternating egg yolk with soy lecithin upgraded post-thaw sperm motility (19%) in cattle [87], but some researchers observed a negative or no impact of soy lecithin on sperm motility [88]. Some researchers have enunciated the conclusive effects of soy lecithin (vegetable origin); egg yolk (animal origin) is still significantly used as a cryoprotectant agent. Liposomes are pure and are used as an alternative to freezing media in horses, pigs, and cattle while sperm cryopreservation [89, 90]. Liposome’s plasma membrane guards the permissibility of water and cryoprotectants [91]. Alcohol, glycerol, and ethylene glycerol are permeable cryoprotectants in cattle sperm cryopreservation [91]. After thawing, glycerol harms sperm quality, depending on the specie [92]. Plasma membrane fluidity is lowered by higher glycerol concentration in thawed swine semen [93]. Elevated concentration of glycerol >3.5%—alters the properties of F-actin (that belongs to globular proteins) in the cytoskeleton [94]. Much research is available exploring substitutes, but glycerol is still considered a sublime cryoprotectant. In research, lower glycerol concentrations were mixed with L-glutamine and trehalose in boar semen [95]. In Pigs, glycerol molecules come in contact with lipid bilayers and change membrane diffusion rates of electrolytes, which results in the osmotic contraction of sperm cells so that sperm can bear low temperatures [96]. Trehalose (60–100 mM) mixed with a low concentration of glycerol (1–3%) upgraded sheep-frozen sperm DNA quality [97]. Glycerol combined with trehalose did not affect cattle sperm quality [98]. In equine, amides cause less harm to sperm during cryopreservation [92]. Changing glycerol with dimethylformamide and methylformamamide increased sperm acrosome integrity, mitochondrial membrane potential, mobility, and capability in horses [99]. In sheep, a combination of 4.7% methyl formamide and 2.3% glycerol increased the integrity and mobility of the plasma membrane [100].

Advertisement

9. Addition of seminal plasma components and other supplements

During semen cryopreservation, seminal plasma is detached and changed with freezing media. In rams, complete or partial removal of seminal plasma downregulates sperm’s freezing capacity [101]. To improve sperm quality, detachment of seminal plasma components is not recommended. In swine frozen-thawed semen, the addition of 5% of seminal plasma before freezing elevated plasma membrane integrity and motility upto 10.5% and 9.2%, respectively [102]. In sheep sperm, the inclusion of 20% seminal plasma upgraded motility (14.7%), plasma membrane integrity (10.4%), and chromatin decondensation (13.9%) [103]. Adding seminal plasma proteins to freezing media has critical functions in sheep [104]. In boar sperm, sperm adhesins attach to glycoproteins in the oviduct, upgrading sperm membrane quality [105]. Furthermore, the task of sperm adhesins is elaborated in sperm cryopreservation. In swine sperm, including seminal plasma proteins AQN-1, AWN, and AQN-3 inhibited the capacitation of sperm at 5°C [106]. BSPs tend to upgrade plasma membrane integrity and halt capacitation [107]. In pigs, adding RSVP14 and RSVP20 in a freezing medium upgraded sperm viability [108]. It explains that sperm quality can be increased or at least sustained by these proteins [109, 110]. In cattle, seminal plasma proteins have a low molecular weight (14–16 kDa) [111]. Adding 1–1.5 mg of protein to MW (14–16 kDa) increased sperm viability by up to 20%. Seminal plasma (SP) proteins of one species can be used as sperm cryopreservation media for other species. SP proteins were added to the freezing medium in cattle [112, 113]. In rams, freezing capacity is elevated after altering bovine serum albumin (BSA) with egg yolk [114, 115]. Adding 4 mg/mL BSA to an excellent medium increased post-thaw sperm quality in bucks [116].

Sperm oxidation is lowered by enzymes inside seminal plasma [117, 118]. Antioxidant defensive enzymes in sperm are catalase, superoxide dismutase (SOD), glutathione reductase, and glutathione peroxidase. Superoxide dismutase is essential in sperm cry tolerance in equids [119]. In various breeds, the protective system of such enzymes is also related to seasonal breeding [120, 121]. In cattle, post-thaw sperm quality increased after adding antioxidants to the freezing medium. Cattle post-thaw sperm viability after inclusion of 100 IU/mL of superoxide dismutase (SOD) [122]. After adding SOD in rams, similar results were seen in post-thaw semen [123]. Post-thaw sperm quality increased after adding glutathione to freezing media in equine and swine, respectively [124, 125]. In rams, adding 2–5 mM glutathione in freezing media increased the acrosome integrity and motility of frozen-thawed sperm [123].

Advertisement

10. Conclusions

Animal biotechnology is now considered a pillar in research and animal production. Its wide application in all animals, including equine, still needs dire attention. Much work has been done in horses but lacks work in donkeys, especially pre- and post-thaw semen quality. Better research and collaborations between scientists, academia, and industry can aid in achieving milestones in equine biotechnology.

Funding

This research was funded by Shandong Province Modern Agricultural Industrial Technology System Project (SDAIT-27), Ministry of Agriculture and Rural Livestock Seed Industry Project “Donkey Camel Species Molecular ID Construction” (19211162), Key research and development project of Shandong Province “Innovation and Demonstration of Key Technologies for Integrated Development of Dong’e Black Donkey Industry” (2021TZXD012).

List of abbreviations

ROSreactive oxygedefn species
mRNAmessenger Ribonucleic Acid
AKTalpha serine/threonine-protein kinase
AWNboar protein
BSPsbinder of sperm (BSP proteins)
miRNAsmicro–ribonucleic acid

References

  1. 1. Akhtar MF, Shafiq M, Ali I. Improving gander reproductive efficacy in the context of globally sustainable goose production. Animals : An Open Access Journal from MDPI. 2021;12:44. DOI: 10.3390/ani12010044
  2. 2. Sherman J. Low temperature research on spermatozoa and eggs. Cryobiology. 1964;1:103-129
  3. 3. Phillips PH, Lardy HA. A yolk-buffer pabulum for the preservation of bull sperm. Journal of Dairy Science. 1940;23:399-404
  4. 4. Polge C, Smith AU, Parkes AS. Revival of spermatozoa after vitrification and dehydration at low temperatures. Nature. 1949;164:666
  5. 5. Loomis P, Graham J. Commercial semen freezing: Individual male variation in cryosurvival and the response of stallion sperm to customized freezing protocols. Animal Reproduction Science. 2008;105:119-128
  6. 6. Salamon S, Maxwell W. Frozen storage of ram semen I. processing, freezing, thawing and fertility after cervical insemination. Animal Reproduction Science. 1995;37:185-249
  7. 7. Nijs M, Creemers E, Cox A, Janssen M, Vanheusden E, Castro-Sanchez Y, et al. Influence of freeze-thawing on hyaluronic acid binding of human spermatozoa. Reproductive BioMedicine Online. 2009;19:202-206
  8. 8. Noda T, Blaha A, Fujihara Y, Gert KR, Emori C, Deneke VE, et al. Sperm membrane proteins DCST1 and DCST2 are required for sperm-egg interaction in mice and fish. Communications Biology. 2022;5:332
  9. 9. Sieme H, Harrison R, Petrunkina A. Cryobiological determinants of frozen semen quality, with special reference to stallion. Animal Reproduction Science. 2008;107:276-292
  10. 10. Morris GJ, Acton E, Murray BJ, Fonseca F. Freezing injury: The special case of the sperm cell. Cryobiology. 2012;64:71-80
  11. 11. Yeste M. Sperm cryopreservation update: Cryodamage, markers, and factors affecting the sperm freezability in pigs. Theriogenology. 2016;85:47-64
  12. 12. Pommer AC, Rutllant J, Meyers SA. The role of osmotic resistance on equine spermatozoal function. Theriogenology. 2002;58:1373-1384
  13. 13. Mazur P, Leibo S, Chu E. A two-factor hypothesis of freezing injury: Evidence from Chinese hamster tissue-culture cells. Experimental Cell Research. 1972;71:345-355
  14. 14. Hammerstedt RH, Graham JK. Cryopreservation of poultry sperm: The enigma of glycerol. Cryobiology. 1992;29:26-38
  15. 15. Brillard J, Bakst M. Quantification of spermatozoa in the sperm-storage tubules of Turkey hens and the relation to sperm numbers in the perivitelline layer of eggs. Biology of Reproduction. 1990;43:271-275
  16. 16. Flores E, Ramió-Lluch L, Bucci D, Fernández-Novell J, Peña A, Rodríguez-Gil J. Freezing-thawing induces alterations in histone H1-DNA binding and the breaking of protein-DNA disulfide bonds in boar sperm. Theriogenology. 2011;76:1450-1464
  17. 17. Ribas-Maynou J, Garcia-Bonavila E, Hidalgo CO, Catalán J, Miró J, Yeste M. Species-specific differences in sperm chromatin decondensation between eutherian mammals underlie distinct lysis requirements. Frontiers in Cell and Development Biology. 2021;9:1143
  18. 18. Gosálvez J, López-Fernández C, Fernández JL, Gouraud A, Holt WV. Relationships between the dynamics of iatrogenic D.N.A. damage and genomic design in mammalian spermatozoa from eleven species. Molecular Reproduction and Development. 2011;78:951-961
  19. 19. McCarthy MJ, Baumber J, Kass PH, Meyers SA. Osmotic stress induces oxidative cell damage to rhesus macaque spermatozoa. Biology of Reproduction. 2010;82:644-651
  20. 20. Kopeika J, Thornhill A, Khalaf Y. The effect of cryopreservation on the genome of gametes and embryos: Principles of cryobiology and critical appraisal of the evidence. Human Reproduction Update. 2015;21:209-227
  21. 21. Stoeckius M, Grün D, Rajewsky N. Paternal RNA contributions in the Caenorhabditis elegans zygote. The EMBO Journal. 2014;33:1740-1750
  22. 22. Kasimanickam V, Kasimanickam R, Arangasamy A, Saberivand A, Stevenson J, Kastelic J. Association between mRNA abundance of functional sperm function proteins and fertility of Holstein bulls. Theriogenology. 2012;78(2007-2019):e2002
  23. 23. Wang P, Wang Y-F, Wang H, Wang C-W, Zan L-S, Hu J-H, et al. HSP90 expression correlation with the freezing resistance of bull sperm. Zygote. 2014;22:239-245
  24. 24. Ortiz-Rodriguez JM, Balao da Silva C, Masot J, Redondo E, Gazquez A, Tapia JA, et al. Rosiglitazone in the thawing medium improves mitochondrial function in stallion spermatozoa through regulating Akt phosphorylation and reduction of caspase 3. PLoS One. 2019;14:e0211994
  25. 25. Urrego R, Rodriguez-Osorio N, Niemann H. Epigenetic disorders and altered gene expression after use of assisted reproductive technologies in domestic cattle. Epigenetics. 2014;9:803-815
  26. 26. Zeng C, Peng W, Ding L, He L, Zhang Y, Fang D, et al. A preliminary study on epigenetic changes during boar spermatozoa cryopreservation. Cryobiology. 2014;69:119-127
  27. 27. Capra E, Turri F, Lazzari B, Cremonesi P, Gliozzi T, Fojadelli I, et al. Sequencing of cryopreserved semen from single bull revealed altered miRNAs and piRNAs expression between high-and low-motile sperm populations. B.M.C. Genomics. 2017;18:1-12
  28. 28. Dai D-H, Qazi IH, Ran M-X, Liang K, Zhang Y, Zhang M, et al. Exploration of miRNA and mRNA profiles in fresh and frozen-thawed boar sperm by transcriptome and small R.N.A. sequencing. International Journal of Molecular Sciences. 2019;20:802
  29. 29. Said TM, Gaglani A, Agarwal A. Implication of apoptosis in sperm cryoinjury. Reproductive Biomedicine Online. 2010;21:456-462
  30. 30. Shangguan A, Zhou H, Sun W, Ding R, Li X, Liu J, et al. Cryopreservation induces alterations of miRNA and mRNA fragment profiles of bull sperm. Frontiers in Genetics. 2020;11:419
  31. 31. Benchaib M, Ajina M, Lornage J, Niveleau A, Durand P, Guérin JF. Quantitation by image analysis of global D.N.A. methylation in human spermatozoa and its prognostic value in in vitro fertilization: A preliminary study. Fertility and Sterility. 2003;80:947-953
  32. 32. Aurich C, Schreiner B, Ille N, Alvarenga M, Scarlet D. Cytosine methylation of sperm D.N.A. in horse semen after cryopreservation. Theriogenology. 2016;86:1347-1352
  33. 33. van der Heijden GW, Ramos L, Baart EB, van den Berg IM, Derijck AA, van der Vlag J, et al. Sperm-derived histones contribute to zygotic chromatin in humans. B.M.C. Developmental Biology. 2008;8:34
  34. 34. Jia G, Fu X, Cheng K, Yue M, Jia B, Hou Y, et al. Spermatozoa cryopreservation alters pronuclear formation and zygotic D.N.A. demethylation in mice. Theriogenology. 2015;83:1000-1006
  35. 35. Ortiz-Rodríguez JM, Martín-Cano FE, Gaitskell-Phillips G, Barrientos AÁ, Rodríguez-Martínez H, Gil MC, et al. Sperm cryopreservation impacts the early development of equine embryos by downregulating specific transcription factors. bioRxiv. 2021. DOI: 10.1101/2021.05.12.443855
  36. 36. Wang M, Gao Y, Qu P, Qing S, Qiao F, Zhang Y, et al. Sperm-borne miR-449b influences cleavage, epigenetic reprogramming and apoptosis of SCNT embryos in bovine. Scientific Reports. 2017;7:13403
  37. 37. Braga D, Setti A, Figueira R, Iaconelli A Jr, Borges E Jr. The negative influence of sperm cryopreservation on the quality and development of the embryo depends on the morphology of the Oocyte. Andrology. 2015;3:723-728
  38. 38. Zhang Y, Dai D, Chang Y, Li Y, Zhang M, Zhou G, et al. Cryopreservation of boar sperm induces differential microRNAs expression. Cryobiology. 2017;76:24-33
  39. 39. Martinez-Cayuela M. Oxygen free radicals and human disease. Biochimie. 1995;77:147-161
  40. 40. Dutta S, Majzoub A, Agarwal A. Oxidative stress and sperm function: A systematic review on evaluation and management. Arab Journal of Urology. 2019;17:87-97
  41. 41. Januskauskas A, Johannisson A, Rodriguez-Martinez H. Subtle membrane changes in cryopreserved bull semen in relation with sperm viability, chromatin structure, and field fertility. Theriogenology. 2003;60:743-758
  42. 42. Flores E, Cifuentes D, Fernández-Novell J, Medrano A, Bonet S, Briz M, et al. Freeze-thawing induces alterations in the protamine-1/DNA overall structure in boar sperm. Theriogenology. 2008;69:1083-1094
  43. 43. Kadirvel G, Kumar S, Kumaresan A. Lipid peroxidation, mitochondrial membrane potential and D.N.A. integrity of spermatozoa in relation to intracellular reactive oxygen species in liquid and frozen-thawed buffalo semen. Animal Reproduction Science. 2009;114:125-134
  44. 44. Rarani FZ, Golshan-Iranpour F, Dashti GR. Correlation between sperm motility and sperm chromatin/D.N.A. damage before and after cryopreservation and the effect of folic acid and nicotinic acid on post-thaw sperm quality in normozoospermic men. Cell and Tissue Banking. 2019;20:367-378
  45. 45. Ugur MR, Saber Abdelrahman A, Evans HC, Gilmore AA, Hitit M, Arifiantini RI, et al. Advances in cryopreservation of bull sperm. Frontiers in Veterinary Science. 2019;6:268
  46. 46. Holt WV, O'Brien J, Abaigar T. Applications and interpretation of computer-assisted sperm analyses and sperm sorting methods in assisted breeding and comparative research. Reproduction, Fertility, and Development. 2007;19:709-718
  47. 47. Yániz JL, Silvestre MA, Santolaria P, Soler C. CASA-Mot in mammals: An update. Reproduction, Fertility, and Development. 2018;30:799-809. DOI: 10.1071/rd17432
  48. 48. Martínez-Pastor F, Tizado EJ, Garde JJ, Anel L, de Paz P. Statistical series: Opportunities and challenges of sperm motility subpopulation analysis. Theriogenology. 2011;75:783-795
  49. 49. Ibanescu I, Siuda M, Bollwein H. Motile sperm subpopulations in bull semen using different clustering approaches–associations with flow cytometric sperm characteristics and fertility. Animal Reproduction Science. 2020;215:106329
  50. 50. Bompart D, García-Molina A, Valverde A, Caldeira C, Yániz J, de Murga MN, et al. CASA-Mot technology: How results are affected by the frame rate and counting chamber. Reproduction, Fertility, and Development. 2018;30:810-819
  51. 51. Soler C, Picazo-Bueno JÁ, Micó V, Valverde A, Bompart D, Blasco FJ, et al. Effect of counting chamber depth on the accuracy of lensless microscopy for the assessment of boar sperm motility. Reproduction, Fertility, and Development. 2018;30:924-934
  52. 52. Khalil WA, El-Harairy MA, Zeidan AE, Hassan MA, Mohey-Elsaeed O. Evaluation of bull spermatozoa during and after cryopreservation: Structural and ultrastructural insights. International Journal of Veterinary Science and Medicine. 2018;6:S49-S56
  53. 53. Roca J, Hernández M, Carvajal G, Vázquez J, Martinez E. Factors influencing boar sperm cryosurvival. Journal of Animal Science. 2006;84:2692-2699
  54. 54. García W, Tabarez A, Palomo MJ. Effect of the type of egg yolk, removal of seminal plasma and donor age on ram sperm cryopreservation. Animal Reproduction (AR). 2018;14:1124-1132
  55. 55. Ferrer M, Canisso I, Ellerbrock R, Podico G, Lister B, Hurley D, et al. Optimization of cryopreservation protocols for cooled-transported stallion semen. Animal Reproduction Science. 2020;221:106581
  56. 56. Rehman A, Ahmad E, Sattar A, Riaz A, Khan JA, Naseer Z, et al. Long term effects of immunization against inhibin on fresh and post-thawed semen quality and sperm kinematics during low and peak breeding seasons in Beetal bucks. Small Ruminant Research. 2021;201:106442
  57. 57. Yánez-Ortiz I, Catalán J, Rodríguez-Gil JE, Miró J, Yeste M. Advances in sperm cryopreservation in farm animals: Cattle, horse, pig and sheep. Animal Reproduction Science. 2021;246:106904. DOI: 10.1016/j.anireprosci.2021.106904
  58. 58. Moura A, Memili E. Functional aspects of seminal plasma and sperm proteins and their potential as molecular markers of fertility. Animal Reproduction (AR). 2018;13:191-199
  59. 59. Mostek A, Dietrich MA, Słowińska M, Ciereszko A. Cryopreservation of bull semen is associated with carbonylation of sperm proteins. Theriogenology. 2017;92:95-102
  60. 60. Riesco FM, Anel-Lopez L, Neila-Montero M, Palacin-Martinez C, Montes-Garrido R, Alvarez M, et al. ProAKAP4 as novel molecular marker of sperm quality in ram: An integrative study in fresh, cooled and cryopreserved sperm. Biomolecules. 2020;10:1046
  61. 61. Zhang X-G, Hu S, Han C, Zhu Q-C, Yan G-J, Hu J-H. Association of heat shock protein 90 with motility of post-thawed sperm in bulls. Cryobiology. 2015;70:164-169
  62. 62. Holt W, Del Valle I, Fazeli A. Heat shock protein A8 stabilizes the bull sperm plasma membrane during cryopreservation: Effects of breed, protein concentration, and mode of use. Theriogenology. 2015;84:693-701
  63. 63. Agre P, King LS, Yasui M, Guggino WB, Ottersen OP, Fujiyoshi Y, et al. Aquaporin water channels–from atomic structure to clinical medicine. The Journal of Physiology. 2002;542:3-16
  64. 64. Yeste M, Morató R, Rodríguez-Gil J, Bonet S, Prieto-Martínez N. Aquaporins in the male reproductive tract and sperm: Functional implications and cryobiology. Reproduction in Domestic Animals. 2017;52:12-27
  65. 65. Vilagran I, Yeste M, Sancho S, Casas I, del Álamo MMR, Bonet S. Relationship of sperm small heat-shock protein 10 and voltage-dependent anion channel 2 with semen freezability in boars. Theriogenology. 2014;82:418-426
  66. 66. Llavanera M, Delgado-Bermúdez A, Fernandez-Fuertes B, Recuero S, Mateo Y, Bonet S, et al. GSTM3, but not IZUMO1, is a cryotolerance marker of boar sperm. Journal of Animal Science and Biotechnology. 2019;10:61
  67. 67. Vilagran I, Yeste M, Sancho S, Castillo J, Oliva R, Bonet S. Comparative analysis of boar seminal plasma proteome from different freezability ejaculates and identification of fibronectin 1 as sperm freezability marker. Andrology. 2015;3:345-356
  68. 68. Rickard J, Leahy T, Soleilhavoup C, Tsikis G, Labas V, Harichaux G, et al. The identification of proteomic markers of sperm freezing resilience in ram seminal plasma. Journal of Proteomics. 2015;126:303-311
  69. 69. AbdelHafez F, Bedaiwy M, El-Nashar SA, Sabanegh E, Desai N. Techniques for cryopreservation of individual or small numbers of human spermatozoa: A systematic review. Human Reproduction Update. 2009;15:153-164
  70. 70. Bergeron A, Manjunath P. New insights towards understanding the mechanisms of sperm protection by egg yolk and milk. Molecular Reproduction and Development. 2006;73:1338-1344
  71. 71. Amirat L, Tainturier D, Jeanneau L, Thorin C, Gérard O, Courtens JL, et al. Bull semen in vitro fertility after cryopreservation using egg yolk LDL: A comparison with Optidyl®, a commercial egg yolk extender. Theriogenology. 2004;61:895-907
  72. 72. Jiang Z-L, Li Q-W, Li W-Y, Hu J-H, Zhao H-W, Zhang S-S. Effect of low density lipoprotein on D.N.A. integrity of freezing–thawing boar sperm by neutral comet assay. Animal Reproduction Science. 2007;99:401-407
  73. 73. Tonieto RA, Goularte K, Gastal GDA, Schiavon RS, Deschamps JC, Lucia T Jr. Cryoprotectant effect of trehalose and low-density lipoprotein in extenders for frozen ram semen. Small Ruminant Research. 2010;93:206-209
  74. 74. Pillet E, Duchamp G, Batellier F, Beaumal V, Anton M, Desherces S, et al. Egg yolk plasma can replace egg yolk in stallion freezing extenders. Theriogenology. 2011;75:105-114
  75. 75. Murphy EM, O'Meara C, Eivers B, Lonergan P, Fair S. Comparison of plant-and egg yolk-based semen diluents on in vitro sperm kinematics and in vivo fertility of frozen-thawed bull semen. Animal Reproduction Science. 2018;191:70-75
  76. 76. Vidal AH, Batista AM, da Silva ECB, Gomes WA, Pelinca MA, Silva SV, et al. Soybean lecithin-based extender as an alternative for goat sperm cryopreservation. Small Ruminant Research. 2013;109:47-51
  77. 77. Aires VA, Hinsch K-D, Mueller-Schloesser F, Bogner K, Mueller-Schloesser S, Hinsch E. In vitro and in vivo comparison of egg yolk-based and soybean lecithin-based extenders for cryopreservation of bovine semen. Theriogenology. 2003;60:269-279
  78. 78. Crespilho A, Sá Filho M, Dell'Aqua J Jr, Nichi M, Monteiro G, Avanzi B, et al. Comparison of in vitro and in vivo fertilizing potential of bovine semen frozen in egg yolk or new lecithin based extenders. Livestock Science. 2012;149:1-6
  79. 79. Luna-Orozco J, González-Ramos M, Calderón-Leyva G, Gaytán-Alemán L, Arellano-Rodríguez F, Ángel-García O, et al. Comparison of different diluents based on liposomes and egg yolk for ram semen cooling and cryopreservation. Iranian Journal of Veterinary Research. 2019;20:126
  80. 80. Medina-León AZ, Domínguez-Mancera B, Cazalez-Penino N, Cervantes-Acosta P, Jácome-Sosa E, Romero-Salas D, et al. Cryopreservation of horse semen with a liposome and trehalose added extender. Austral Journal of Veterinary Sciences. 2019;51:119-123
  81. 81. Röpke T, Oldenhof H, Leiding C, Sieme H, Bollwein H, Wolkers W. Liposomes for cryopreservation of bovine sperm. Theriogenology. 2011;76:1465-1472
  82. 82. Alvarenga MA, Papa FO, Landim-Alvarenga F, Medeiros A. Amides as cryoprotectants for freezing stallion semen: A review. Animal Reproduction Science. 2005;89:105-113
  83. 83. Buhr MM, Fiser P, Bailey JL, Curtis EF. Cryopreservation in different concentrations of glycerol alters boar sperm and their membranes. Journal of Andrology. 2001;22:961-969
  84. 84. Macías García B, Ortega Ferrusola C, Aparicio I, Miró-Morán A, Morillo Rodriguez A, Gallardo Bolanos J, et al. Toxicity of glycerol for the stallion spermatozoa: Effects on membrane integrity and cytoskeleton, lipid peroxidation and mitochondrial membrane potential. Theriogenology. 2012;77:1280-1289
  85. 85. De Mercado E, Hernandez M, Sanz E, Rodriguez A, Gomez E, Vazquez J, et al. Evaluation of l-glutamine for cryopreservation of boar spermatozoa. Animal Reproduction Science. 2009;115:149-157
  86. 86. Gutiérrez-Pérez O, de Lourdes Juárez-Mosqueda M, Carvajal SU, Ortega MET. Boar spermatozoa cryopreservation in low glycerol/trehalose enriched freezing media improves cellular integrity. Cryobiology. 2009;58:287-292
  87. 87. Özmen MF, Cirit Ü, Arıcı R, Demir K, Kurt D, Pabuccuoğlu S, et al. Evaluation of synergic effects of iodixanol and trehalose on cryosurvival of electroejaculated ram semen. Andrologia. 2020;52:e13656
  88. 88. Büyükleblebici S, Tuncer PB, Bucak MN, Eken A, Sarıözkan S, Taşdemir U, et al. Cryopreservation of bull sperm: Effects of extender supplemented with different cryoprotectants and antioxidants on sperm motility, antioxidant capacity and fertility results. Animal Reproduction Science. 2014;150:77-83
  89. 89. Wu Z, Zheng X, Luo Y, Huo F, Dong H, Zhang G, et al. Cryopreservation of stallion spermatozoa using different cryoprotectants and combinations of cryoprotectants. Animal Reproduction Science. 2015;163:75-81
  90. 90. Forero-Gonzalez R, Celeghini E, Raphael C, Andrade A, Bressan F, Arruda R. Effects of bovine sperm cryopreservation using different freezing techniques and cryoprotective agents on plasma, acrosomal and mitochondrial membranes. Andrologia. 2012;44:154-159
  91. 91. Ashworth P, Harrison R, Miller N, Plummer J, Watson P. Survival of ram spermatozoa at high dilution: Protective effect of simple constituents of culture media as compared with seminal plasma. Reproduction, Fertility, and Development. 1994;6:173-180
  92. 92. Hernández M, Roca J, Calvete JJ, Sanz L, Muiño-Blanco T, Cebrián-Pérez JA, et al. Cryosurvival and in vitro fertilizing capacity postthaw is improved when boar spermatozoa are frozen in the presence of seminal plasma from good freezer boars. Journal of Andrology. 2007;28:689-697. DOI: 10.2164/jandrol.107.002725
  93. 93. Ramírez-Vasquez R, Cesari A, Greco MB, Cano A, Hozbor F. Extenders modify the seminal plasma ability to minimize freeze-thaw damage on ram sperm. Reproduction in Domestic Animals. 2019;54:1621-1629. DOI: 10.1111/rda.13571
  94. 94. Rodríguez-Martínez H, Kvist U, Ernerudh J, Sanz L, Calvete JJ. Seminal plasma proteins: What role do they play? American Journal of Reproductive Immunology. 2011;66(Suppl 1):11-22. DOI: 10.1111/j.1600-0897.2011.01033.x
  95. 95. Dostàlovà Z, Calvete JJ, Töpfer-Petersen E. Interaction of non-aggregated boar AWN-1 and AQN-3 with phospholipid matrices. A model for coating of spermadhesins to the sperm surface. Biological Chemistry Hoppe-Seyler. 1995;376:237-242
  96. 96. Vadnais ML, Roberts KP. Seminal plasma proteins inhibit in vitro-and cooling-induced capacitation in boar spermatozoa. Reproduction, Fertility, and Development. 2010;22:893-900
  97. 97. Manjunath P, Bergeron A, Lefebvre J, Fan J. Seminal plasma proteins: Functions and interaction with protective agents during semen preservation. Society of Reproduction and Fertility Supplement. 2007;65:217
  98. 98. Muiño-Blanco T, Pérez-Pé R, Cebrián-Pérez J. Seminal plasma proteins and sperm resistance to stress. Reproduction in Domestic Animals. 2008;43:18-31
  99. 99. Barrios B, Fernández-Juan M, Muiño-Blanco T, Cebrián-Pérez JA. Immunocytochemical localization and biochemical characterization of two seminal plasma proteins that protect ram spermatozoa against cold shock. Journal of Andrology. 2005;26:539-549
  100. 100. Bernardini A, Hozbor F, Sanchez E, Fornes MW, Alberio R, Cesari A. Conserved ram seminal plasma proteins bind to the sperm membrane and repair cryopreservation damage. Theriogenology. 2011;76:436-447
  101. 101. Rueda F, Garcés T, Herrera R, Arbeláez L, Peña M, Velásquez H, et al. Las proteínas del plasma seminal incrementan la viabilidad espermática post-descongelación del semen de toros Sanmartinero. Revista MVZ Córdoba. 2013;18:3327-3335
  102. 102. Susilowati S, Triana IN, Sardjito T, Suprayogi TW, Wurlina W, Mustofa I. Effect of Simmental bull seminal plasma protein in egg yolk-citrate extender on Kacang buck semen fertility. Cryobiology. 2020;97:20-27
  103. 103. Uysal O, Bucak M. Effects of oxidized glutathione, bovine serum albumin, cysteine and lycopene on the quality of frozen-thawed ram semen. Acta Veterinaria. 2007;76:383-390
  104. 104. Matsuoka T, Imai H, Kohno H, Fukui Y. Effects of bovine serum albumin and trehalose in semen diluents for improvement of frozen-thawed ram spermatozoa. The Journal of Reproduction and Development. 2006;52:675-683
  105. 105. Fukui Y, Kohno H, Togari T, Hiwasa M. Fertility of ewes inseminated intrauterinally with frozen semen using extender containing bovine serum albumin. Journal of Reproduction and Development. 2007;53:959-962
  106. 106. Kaewkesa T, Sathanawongs A, Oranratnachai A, Sumretprasong J. The goat semen quality after being frozen using albumin and cholesterol substituted for egg yolk in semen extender. The Thai Veterinary Medicine. 2016;46:201
  107. 107. Aitken RJ. Free radicals, lipid peroxidation and sperm function. Reproduction, Fertility, and Development. 1995;7:659-668
  108. 108. Ortega-Ferrusola C, Martin Muñoz P, Ortiz-Rodriguez JM, Anel-López L, Balao da Silva C, Álvarez M, et al. Depletion of thiols leads to redox deregulation, production of 4-hydroxinonenal and sperm senescence: A possible role for G.S.H. regulation in spermatozoa. Biology of Reproduction. 2019;100:1090-1107
  109. 109. Papas M, Catalán J, Fernandez-Fuertes B, Arroyo L, Bassols A, Miró J, et al. Specific activity of superoxide dismutase in stallion seminal plasma is related to sperm cryotolerance. Antioxidants. 2019;8:539
  110. 110. Marti E, Mara L, Marti J, Muiño-Blanco T, Cebrián-Pérez J. Seasonal variations in antioxidant enzyme activity in ram seminal plasma. Theriogenology. 2007;67:1446-1454
  111. 111. Papas M, Arroyo L, Bassols A, Catalán J, Bonilla-Correal S, Gacem S, et al. Activities of antioxidant seminal plasma enzymes (S.O.D., C.a.T., G.P.X. and G.S.R.) are higher in jackasses than in stallions and are correlated with sperm motility in jackasses. Theriogenology. 2019;140:180-187
  112. 112. Olfati Karaji R, Daghigh Kia H, Ashrafi I. Effects of in combination antioxidant supplementation on microscopic and oxidative parameters of freeze–thaw bull sperm. Cell and Tissue Banking. 2014;15:461-470
  113. 113. Silva S, Soares A, Batista A, Almeida F, Nunes J, Peixoto C, et al. In vitro and in vivo evaluation of ram sperm frozen in tris egg-yolk and supplemented with superoxide dismutase and reduced glutathione. Reproduction in Domestic Animals. 2011;46:874-881
  114. 114. de Oliveira RA, Wolf CA, de Oliveira Viu MA, Gambarini ML. Addition of glutathione to an extender for frozen equine semen. Journal of Equine Veterinary Science. 2013;33:1148-1152
  115. 115. Gadea J, García-Vazquez F, Matás C, Gardón JC, Cánovas S, Gumbao D. Cooling and freezing of boar spermatozoa: Supplementation of the freezing media with reduced glutathione preserves sperm function. Journal of Andrology. 2005;26:396-404
  116. 116. Al-Mutary MG. Use of antioxidants to augment semen efficiency during liquid storage and cryopreservation in livestock animals: A review. Journal of King Saud University - Science. 2021;33:101226
  117. 117. Guthrie H, Welch G. Effects of reactive oxygen species on sperm function. Theriogenology. 2012;78:1700-1708
  118. 118. Treulen F, Aguila L, Arias ME, Jofré I, Felmer R. Impact of post-thaw supplementation of semen extender with antioxidants on the quality and function variables of stallion spermatozoa. Animal Reproduction Science. 2019;201:71-83
  119. 119. Banday MN, Lone FA, Rasool F, Rashid M, Shikari A. Use of antioxidants reduce lipid peroxidation and improve quality of crossbred ram sperm during its cryopreservation. Cryobiology. 2017;74:25-30
  120. 120. de Souza CV, Brandão FZ, Santos JDR, Alfradique VAP, Dos Santos VMB, da Cruz Morais MC, et al. Effect of different concentrations of L-carnitine in extender for semen cryopreservation in sheep. Cryobiology. 2019;89:104-108
  121. 121. Del Valle I, Souter A, Maxwell W, Muino-Blanco T, Cebrián-Pérez J. Function of ram spermatozoa frozen in diluents supplemented with casein and vegetable oils. Animal Reproduction Science. 2013;138:213-219
  122. 122. Carrera-Chávez JM, Jiménez-Aguilar EE, Acosta-Pérez TP, Núñez-Gastélum JA, Quezada-Casasola A, Escárcega-Ávila AM, et al. Effect of Moringa oleifera seed extract on antioxidant activity and sperm characteristics in cryopreserved ram semen. Journal of Applied Animal Research. 2020;48:114-120
  123. 123. Motlagh MK, Sharafi M, Zhandi M, Mohammadi-Sangcheshmeh A, Shakeri M, Soleimani M, et al. Antioxidant effect of rosemary (Rosmarinus officinalis L.) extract in soybean lecithin-based semen extender following freeze–thawing process of ram sperm. Cryobiology. 2014;69:217-222
  124. 124. Nouri H, Shojaeian K, Samadian F, Lee S, Kohram H, Lee JI. Using resveratrol and epigallocatechin-3-gallate to improve cryopreservation of stallion spermatozoa with low quality. Journal of Equine Veterinary Science. 2018;70:18-25
  125. 125. Zhu Z, Li R, Fan X, Lv Y, Zheng Y, Hoque S, et al. Resveratrol improves boar sperm quality via 5AMP-activated protein kinase activation during cryopreservation. Oxidative Medicine and Cellular Longevity. 2019;2019:15

Written By

Muhammad Faheem Akhtar and Changfa Wang

Submitted: 13 December 2022 Reviewed: 15 December 2022 Published: 16 May 2023